Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS Pathog ; 20(3): e1012095, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38512979

RESUMEN

The 1858C>T allele of the tyrosine phosphatase PTPN22 is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its pro-autoimmune allele has in anti-viral immunity remains poorly defined. Here, we use single cell RNA-sequencing and functional studies to interrogate the impact of this pro-autoimmune allele on anti-viral immunity during Lymphocytic Choriomeningitis Virus clone 13 (LCMV-cl13) infection. Mice homozygous for this allele (PEP-619WW) clear the LCMV-cl13 virus whereas wildtype (PEP-WT) mice cannot. This is associated with enhanced anti-viral CD4 T cell responses and a more immunostimulatory CD8α- cDC phenotype. Adoptive transfer studies demonstrated that PEP-619WW enhanced anti-viral CD4 T cell function through virus-specific CD4 T cell intrinsic and extrinsic mechanisms. Taken together, our data show that the pro-autoimmune allele of Ptpn22 drives a beneficial anti-viral immune response thereby preventing what is normally a chronic virus infection.


Asunto(s)
Enfermedades Autoinmunes , Coriomeningitis Linfocítica , Animales , Ratones , Alelos , Enfermedades Autoinmunes/genética , Autoinmunidad/genética , Monoéster Fosfórico Hidrolasas/genética , Tirosina
2.
J Immunol ; 207(6): 1662-1671, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34417261

RESUMEN

The 1858C>T allele of the tyrosine phosphatase PTPN22 (causing amino acid substitution R620W in encoded protein lymphoid tyrosine phosphatase) is present in 5-10% of the North American population and is strongly associated with numerous autoimmune diseases. Although much research has been done to define how this allele potentiates autoimmunity, the influence PTPN22 and its proautoimmune allele have in tumor immunity is poorly defined. To interrogate the role this allele may have in the antitumor immune response, we used CRISPR/Cas9 to generate mice in which the ortholog of lymphoid tyrosine phosphatase, PEST domain-enriched protein (PEP), is mutated at position 619 to produce the relevant proautoimmune mutation (R619W). Results of this study show that mice homozygous for this alteration (PEP-619WW) resist tumor growth as compared with wild-type mice. Consistent with these results, tumors from PEP-619WW mice have more CD45 infiltrates containing more activated CD8 T cells and CD4 T cells. In addition, there are more conventional dendritic cell type 1 (cDC1) cells and fewer myeloid-derived suppressor cells in tumors from PEP-619WW mice. Interestingly, the tumor-infiltrating PEP-619WW cDC1 cells have decreased PD-L1 expression compared with cDC1 cells from PEP-wild-type mice. Taken together, our data show that the proautoimmune allele of Ptpn22 drives a strong antitumor response in innate and adaptive immune cells resulting in superior control of tumors.


Asunto(s)
Alelos , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/inmunología , Inmunidad , Melanoma Experimental/enzimología , Melanoma Experimental/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/inmunología , Animales , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Femenino , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias/métodos , Fenotipo , Neoplasias Cutáneas/patología
3.
J Immunol ; 199(8): 2713-2720, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28864471

RESUMEN

Peripheral tolerance mechanisms exist to prevent autoimmune destruction by self-reactive T cells that escape thymic deletion. Dominant tolerance imposed by CD4+Foxp3+ T regulatory cells can actively control autoaggressive T cell responses. Tolerance mechanisms that act endogenous to the T cell also exist. These mechanisms include T cell inactivation (anergy) and deletion. A major difference between anergic T cells and T cells undergoing peripheral deletion is the capacity of the latter to still signal through MAPKs upon TCR stimulation, suggesting these signals may be required for T deletion. In this study, we used several different models of CD8 T cell deletion to investigate the contribution of MAPK activation. Using chemical inhibitors, we established that inhibition of p38, but not ERK or JNK, rescue T cells from undergoing peripheral deletion both in vitro and in vivo. Using T cell-specific murine lines genetically altered in expression of p38α, and mice in which p38α was deleted only in CD11c-expressing cells, we surprisingly found that CD8 T cell-intrinsic p38α activation was not responsible for increased survival, but rather that inhibition of p38α in the Ag-presenting dendritic cells prevented CD8 T cell deletion.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Supresión Clonal , Células Dendríticas/inmunología , Tolerancia Periférica , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antígeno CD11c/metabolismo , Línea Celular , Reactividad Cruzada , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética
4.
Proc Natl Acad Sci U S A ; 113(46): E7231-E7239, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799548

RESUMEN

The protein encoded by the autoimmune-associated protein tyrosine phosphatase nonreceptor type 22 gene, PTPN22, has wide-ranging effects in immune cells including suppression of T-cell receptor signaling and promoting efficient production of type I interferons (IFN-I) by myeloid cells. Here we show that mice deficient in PTPN22 resist chronic viral infection with lymphocytic choriomeningitis virus clone 13 (LCMV cl13). The numbers and function of viral-specific CD4 T lymphocytes is greatly enhanced, whereas expression of the IFNß-induced IL-2 repressor, cAMP-responsive element modulator (CREM) is reduced. Reduction of CREM expression in wild-type CD4 T lymphocytes prevents the loss of IL-2 production by CD4 T lymphocytes during infection with LCMV cl13. These findings implicate the IFNß/CREM/IL-2 axis in regulating T-lymphocyte function during chronic viral infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/inmunología , Animales , Modulador del Elemento de Respuesta al AMP Cíclico/inmunología , Interferón Tipo I/inmunología , Interleucina-10/inmunología , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética
5.
Eur J Immunol ; 47(3): 575-584, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28083937

RESUMEN

Secreted microvesicles (MVs) are potent inflammatory triggers that stimulate autoreactive B and T cells, causing Type 1 Diabetes in non-obese diabetic (NOD) mice. Proteomic analysis of purified MVs released from islet cells detected the presence of endogenous retrovirus (ERV) antigens, including Env and Gag sequences similar to the well-characterized murine leukemia retroviruses. This raises the possibility that ERV antigens may be expressed in the pancreatic islets via MV secretion. Using virus-like particles produced by co-expressing ERV Env and Gag antigens, and a recombinant gp70 Env protein, we demonstrated that NOD but not diabetes-resistant mice developed anti-Env autoantibodies that increase in titer as disease progresses. A lentiviral-based RNA interference knockdown of Gag revealed that Gag contributes to the MV-induced T-cell response, whose diabetogenic function can be demonstrated via cell-transfer into immune-deficient mice. Finally, we observed that Gag and Env are expressed in NOD islet-derived primary mesenchymal stem cells (MSCs). However, MSCs derived from the islets of diabetes-resistant mice do not express the antigens. Taken together, abnormal ERV activation and secretion of MVs may induce anti-retroviral responses to trigger autoimmunity.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Retrovirus Endógenos/inmunología , Productos del Gen env/metabolismo , Productos del Gen gag/metabolismo , Islotes Pancreáticos/inmunología , Células Madre Mesenquimatosas/metabolismo , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Autoanticuerpos/sangre , Autoinmunidad , Micropartículas Derivadas de Células/inmunología , Células Cultivadas , Femenino , Productos del Gen env/genética , Productos del Gen gag/genética , Humanos , Islotes Pancreáticos/metabolismo , Activación de Linfocitos , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , ARN Interferente Pequeño/genética , Linfocitos T/trasplante
6.
Curr Diab Rep ; 17(12): 130, 2017 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-29080983

RESUMEN

PURPOSE OF REVIEW: The initial autoimmune trigger of type 1 diabetes (T1D) remains unclear. In non-obese diabetic (NOD) mice, islet inflammation starts early in life, suggesting the presence of an endogenous trigger for the spontaneous autoimmune response in this T1D mouse model. In this review, we argue that abnormal release of exosomes might be the trigger of the early inflammatory and autoimmune responses in the islets. RECENT FINDINGS: Exosomes are nano-sized membrane complexes that are secreted by cells following fusion of late endosomes and/or multivesicular bodies with the plasma membrane. They are known extracellular messengers, communicating among neighboring cells via transporting large molecules from parent cells to recipient cells. Recent evidence demonstrates that these extracellular vesicles can modulate immune responses. It has been shown that insulinoma and islet mesenchymal stem cell-released exosomes are potent immune stimuli that can induce autoreactive B and T cells. Searching for candidate antigens in the exosomes identified endogenous retrovirus (ERV) Env and Gag antigens, which are homologous to an endogenous murine leukemia retrovirus. Autoantibodies and autoreactive T cells spontaneously developed in NOD mice can react to these retroviral antigens. More importantly, expression of the retroviral antigens in the islet mesenchymal stem cells is associated with disease susceptibility, and the expression is restricted to T1D-susceptible but not resistant mouse strains. Exosomes are novel autoimmune targets, carrying autoantigens that can stimulate innate and adaptive immune responses. An abnormal or excess release of exosomes, particularly those ones containing endogenous retroviral antigens might be responsible for triggering tissue-specific inflammatory and autoimmune responses.


Asunto(s)
Antígenos/inmunología , Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Exosomas/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos NOD , Linfocitos T/inmunología
7.
J Immunol ; 192(4): 1415-24, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24453256

RESUMEN

A single nucleotide polymorphism in PTPN22 (R620W), which encodes the Lyp tyrosine phosphatase, has been linked to a number of autoimmune diseases including type 1 diabetes, rheumatoid arthritis, and systemic lupus erythematosus. Studies in PTPN22 knockout (KO) mice and in mice expressing the mouse homolog of the pro-autoimmune allele, PEP(R619W), have reported increased germinal center activity and enhanced Ab production. In this article, we present findings that explain the basis for increased germinal center activity in PTPN22 mutant mice. As compared with their wild type equivalents, T follicular helper cells from PTPN22 KO mice proliferate and accumulate to a greater extent, and exhibit enhanced production of IL-21. The follicular regulatory T cells in PTPN22 KO mice do not expand to effectively regulate these T follicular helper cells, resulting in an increase in B cell numbers and Ab production. This is evident in the KBxN mouse model of arthritis in which PTPN22 deficiency results in increased severity of disease. Our findings demonstrate the importance of cell type-specific PTPN22 activity on regulation of Ab production.


Asunto(s)
Artritis/inmunología , Centro Germinal/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Formación de Anticuerpos/genética , Artritis/genética , Autoinmunidad/inmunología , Linfocitos B/inmunología , Proliferación Celular , Fibronectinas , Predisposición Genética a la Enfermedad , Interleucinas/biosíntesis , Interleucinas/metabolismo , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Proteína Tirosina Fosfatasa no Receptora Tipo 22/deficiencia , Proteínas/metabolismo
8.
J Immunol ; 193(7): 3409-16, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25156361

RESUMEN

Peripheral tolerance mechanisms are in place to prevent T cells from mediating aberrant immune responses directed against self and environmental Ags. Mechanisms involved in the induction of peripheral tolerance include T cell-intrinsic pathways, such as anergy or deletion, or exogenous tolerance mediated by regulatory T cells. We have previously shown that the density of peptide-MHC class I recognized by the TCR determines whether CD8(+) T cells undergo anergy or deletion. Specifically, using a TCR-transgenic CD8(+) T cell model, we demonstrated that persistent peripheral exposure to low- or high-dose peptides in the absence of inflammatory signals resulted in clonal deletion or anergy of the T cell, respectively. In this study, by altering the affinity of the peptide-MHC tolerogen for TCR, we have confirmed that this mechanism is dependent on the level of TCR signaling that the CD8(+) T cell receives. Using altered peptide ligands (APLs) displaying high TCR affinities, we show that increasing the TCR signaling favors anergy induction. Conversely, using APLs displaying a decreased TCR affinity tilted our system in the direction of deletional tolerance. We demonstrate how differential peripheral CD8(+) T cell tolerance mechanisms are controlled by both the potency and density of MHC class I-peptide tolerogen.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Tolerancia Inmunológica/fisiología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Antígenos/genética , Linfocitos T CD8-positivos/citología , Antígenos de Histocompatibilidad Clase I/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Péptidos/genética , Receptores de Antígenos de Linfocitos T/genética
9.
Clin Immunol ; 156(1): 65-73, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25463433

RESUMEN

A single nucleotide polymorphism in PTPN22 is linked to increased disease susceptibility in a range of autoimmune diseases including systemic lupus erythematosus (SLE). PTPN22 encodes the Lyp phosphatase that dampens TCR signaling and is necessary for signaling downstream of toll-like receptors in myeloid cells. To understand these dual functions in disease, we examined the impact of deficiency in PTPN22 on a spontaneous murine model of SLE. Male PTPN22 KO mice carrying BXSB chromosome 1 and the Yaa disease accelerating factor developed disease at a similar rate and severity as PTPN22 WT. In contrast, although female mice showed no differences in survival in the absence of PTPN22, autoantibody production was significantly increased and splenic populations associated with pathogenesis in this model were expanded in the PTPN22 KO group. These findings support the notion that when coupled with other predisposing autoimmunity genes, PTPN22 deficiency contributes to a predisposition to lupus pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Animales , Femenino , Citometría de Flujo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Ratones , Ratones Noqueados
10.
Science ; 384(6702): eade8520, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38900864

RESUMEN

Unleashing antitumor T cell activity by checkpoint inhibitor immunotherapy is effective in cancer patients, but clinical responses are limited. Cytokine signaling through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway correlates with checkpoint immunotherapy resistance. We report a phase I clinical trial of the JAK inhibitor ruxolitinib with anti-PD-1 antibody nivolumab in Hodgkin lymphoma patients relapsed or refractory following checkpoint inhibitor immunotherapy. The combination yielded a best overall response rate of 53% (10/19). Ruxolitinib significantly reduced neutrophil-to-lymphocyte ratios and percentages of myeloid suppressor cells but increased numbers of cytokine-producing T cells. Ruxolitinib rescued the function of exhausted T cells and enhanced the efficacy of immune checkpoint blockade in preclinical solid tumor and lymphoma models. This synergy was characterized by a switch from suppressive to immunostimulatory myeloid cells, which enhanced T cell division.


Asunto(s)
Enfermedad de Hodgkin , Inhibidores de Puntos de Control Inmunológico , Inhibidores de las Cinasas Janus , Nitrilos , Nivolumab , Pirazoles , Pirimidinas , Linfocitos T , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sinergismo Farmacológico , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/terapia , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Quinasas Janus/antagonistas & inhibidores , Nitrilos/uso terapéutico , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Linfocitos T/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C
11.
Proc Natl Acad Sci U S A ; 105(43): 16683-8, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18936481

RESUMEN

Because of mechanisms of self-tolerance, many tumor-specific CD8 T cells exhibit low avidity for tumor antigens and would benefit from strategies that enhance their numbers and effector function. Here we demonstrate that the combined use of two different types of immune adjuvants, one that directly targets the CD8 cell, IL-2/anti-IL-2 mAb complexes, and one that targets the innate immune system, poly(I:C), can achieve this goal. Provision of IL-2/mAb complexes was found to enhance the activation and effector function of low-avidity tumor-specific T cells, yet this was insufficient to achieve tumor eradication. The addition of poly(I:C) further increased the accumulation of granzyme B-expressing effectors within the tumor and resulted in tumor eradication. This strategy presents many of the benefits of whole-body irradiation, including the provision of high levels of homeostatic cytokines, enhanced expansion of effector cells relative to regulatory T cells, and provision of inflammatory cytokines, and is therefore likely to serve as a strategy for both tumor vaccines and adoptive immunotherapy of cancer.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Inmunidad/efectos de los fármacos , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Citocinas , Inmunoterapia Adoptiva/métodos , Interleucina-2/inmunología , Ratones , Ratones Transgénicos , Poli I-C/uso terapéutico , Resultado del Tratamiento
12.
J Exp Med ; 196(3): 323-33, 2002 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12163561

RESUMEN

Professional antigen-presenting cells (APCs) are capable of transporting self-antigens from peripheral tissues to secondary lymphoid organs where they are presented to potentially autoreactive CD8(+) T cells. In the absence of an inflammatory response, this results in immune tolerance. The presence of activated, antigen-specific CD4(+) T cells converts this tolerogenic encounter into an immunogenic one by promoting extensive proliferation of CD8(+) T cells and their development into effectors. Surprisingly, activation of APCs with an agonistic antibody specific for CD40 could not substitute for CD4(+) help in this task. Anti-CD40 induced recruitment of dendritic cells expressing high levels of B7 costimulatory molecules into the lymph nodes, which in turn, greatly enhanced activation and expansion of CD8(+) T cells. However, these activated CD8(+) cells did not demonstrate effector function. We conclude that proliferative potential and gain of effector function are separable events in the differentiation program of CD8(+) T cells.


Asunto(s)
Autoantígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos , Animales , Antígeno B7-1/fisiología , Linfocitos T CD4-Positivos/fisiología , Antígenos CD40/fisiología , Diabetes Mellitus/etiología , Tolerancia Inmunológica , Interferón gamma/biosíntesis , Interleucina-12/farmacología , Ratones , Ratones Endogámicos BALB C , Receptores de Antígenos de Linfocitos T/fisiología
13.
Immunol Lett ; 223: 62-70, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32335144

RESUMEN

Endogenous retrovirus (ERV) are remnants of ancient retroviruses that have been incorporated into the genome and evidence suggests that they may play a role in the etiology of T1D. We previously identified a murine leukemia retrovirus-like ERV whose Env and Gag antigens are involved in autoimmune responses in non-obese diabetic (NOD) mice. In this study, we show that the Gag antigen is present in the islet stromal cells. Although Gag gene transcripts were present, Gag protein was not detected in diabetes-resistant mice. Cloning and sequencing analysis of individual Gag genes revealed that NOD islets express Gag gene variants with complete open-reading frames (ORFs), in contrast to the diabetes-resistant mice, whose islet Gag gene transcripts are mostly non-ORFs. Importantly, the ORFs obtained from the NOD islets are extremely heterogenous, coding for various mutants that are absence in the genome. We further show that Gag antigens are stimulatory for autoreactive T cells and identified one islet-expressing Gag variant that contains an altered peptide ligand capable of inducing IFN-gamma release by the T cells. The data highlight a unique retrovirus-like factor in the islets of the NOD mouse strain, which may participate in key events triggering autoimmunity and T1D.


Asunto(s)
Autoantígenos/metabolismo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Retrovirus Endógenos/fisiología , Productos del Gen gag/metabolismo , Islotes Pancreáticos/metabolismo , Linfocitos T/inmunología , Animales , Autoantígenos/inmunología , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Productos del Gen gag/inmunología , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD
14.
Diabetes ; 65(8): 2134-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27207523

RESUMEN

An allelic variant of protein tyrosine phosphatase nonreceptor type 22 (PTPN22), PTPN22(R620W), is strongly associated with type 1 diabetes (T1D) in humans and increases the risk of T1D by two- to fourfold. The NOD mouse is a spontaneous T1D model that shares with humans many genetic pathways contributing to T1D. We hypothesized that the introduction of the murine orthologous Ptpn22(R619W) mutation to the NOD genome would enhance the spontaneous development of T1D. We microinjected CRISPR-Cas9 and a homology-directed repair template into NOD single-cell zygotes to introduce the Ptpn22(R619W) mutation to its endogenous locus. The resulting Ptpn22(R619W) mice showed increased insulin autoantibodies and earlier onset and higher penetrance of T1D. This is the first report demonstrating enhanced T1D in a mouse modeling human PTPN22(R620W) and the utility of CRISPR-Cas9 for direct genetic alternation of NOD mice.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Alelos , Animales , Western Blotting , Sistemas CRISPR-Cas/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Genoma/genética , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Polimorfismo de Nucleótido Simple/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Oncoimmunology ; 1(8): 1239-1247, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23243587

RESUMEN

Weak T-cell antigen receptor (TCR)-ligand interactions are sufficient to activate naïve CD8(+) T cells, but generally do not result in tumor eradication. How differences in TCR affinity affect the regulation of T-cell function in an immunosuppressive tumor environment has not been investigated. We have examined the functional differences of high- vs. low-affinity CD8(+) T cells and we observed that infiltration, accumulation, survival and cytotoxicity within the tumor are severely impacted by the strength of TCR-ligand interactions. In addition, high-affinity CD8(+) T cells were found to exhibit lower expression of inhibitory molecules including PD-1, LAG-3 and NKG2A, thus being less susceptible to suppressive mechanisms. Interferon γ and autocrine interleukin-2 were both found to influence the level of expression of these molecules. Interestingly, although high-affinity CD8(+) T cells were superior to low-affinity CD8(+) T cells in their ability to effect tumor eradication, they could be further improved by the presence of tumor specific CD4(+) T cells. These findings illustrate the importance of both TCR affinity and tumor-specific CD4 help in tumor immunotherapy.

16.
J Immunol ; 175(10): 6615-23, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16272316

RESUMEN

Under noninflammatory conditions, both naive and central memory CD8 T cells can be eliminated in the periphery with either soluble peptide or cross-presented Ag. Here, we assess the tolerance susceptibility of tissue-resident memory CD8 T cells in mice to these two forms of tolerogen. Soluble peptide specifically eliminated the majority of memory CD8 cells present in both lymphoid and extralymphoid tissues including lung and liver, but was unable to reduce the number present in the CNS. In contrast, systemic cross-presentation of Ag by dendritic cells resulted in successful elimination of memory cells only from the spleen, with no significant reduction in the numbers of tissue-resident memory cells in the lung. The fact that tissue-resident memory cells were unable to access cross-presented Ag suggests that either the memory cells in the lung do not freely circulate out of the tissue, or that they circulate through a region in the spleen devoid of cross-presented Ag. Thus, although tissue-resident memory cells are highly susceptible to tolerance induction, both the form of tolerogen and location of the T cells can determine their accessibility to tolerogen and the degree to which they are successfully deleted from specific tissues.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Animales , Antígenos Virales , Reacciones Cruzadas , Femenino , Tolerancia Inmunológica , Virus de la Influenza A/inmunología , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas de la Nucleocápside , Nucleoproteínas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas de Unión al ARN/inmunología , Solubilidad , Linfocitos T Citotóxicos/inmunología , Proteínas del Núcleo Viral/inmunología
17.
J Immunol ; 174(5): 2563-72, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728462

RESUMEN

A major challenge in tumor immunology is how best to activate the relatively low avidity self-specific and tumor-specific T cells that are available in the self-tolerant repertoire. To address this issue, we produced a TCR transgenic mouse expressing a class I-restricted hemagglutinin (HA)-specific TCR (clone 1 TCR) derived from a mouse that expressed HA as a self-Ag in the insulin-producing beta cells of the pancreatic islets (InsHA) mice. Upon transfer of clone 1 TCR CD8(+) T cells into InsHA mice, very few cells were activated by cross-presented HA, indicating that the cells were retained in InsHA mice because they ignored the presence of Ag, and not because they were functionally inactivated by anergy or tuning. Upon transfer into recipient mice in which HA is expressed at high concentrations as a tumor-associated Ag in spontaneously arising insulinomas (RIP-Tag2-HA mice), a high proportion of clone 1 cells were activated when they encountered cross-presented tumor Ag in the pancreatic lymph nodes. However, the activated cells exhibited very weak effector function and were soon tolerized. The few activated cells that did migrate to the tumor were unable to delay tumor progression. However, when HA-specific CD4 helper cells were cotransferred with clone 1 cells into RIP-Tag2-HA recipients and the mice were vaccinated with influenza, clone 1 cells were found to exert a significant level of effector function and could delay tumor growth. This tumor model should prove of great value in identifying protocols that can optimize the function of low avidity tumor-specific T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Insulinoma/inmunología , Neoplasias Pancreáticas/inmunología , Animales , Antígenos de Neoplasias/metabolismo , Autoantígenos/biosíntesis , Autoantígenos/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Adhesión Celular/genética , Adhesión Celular/inmunología , Diferenciación Celular/inmunología , Células Clonales , Reactividad Cruzada/genética , Citotoxicidad Inmunológica/genética , Epítopos de Linfocito T/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/biosíntesis , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Insulinoma/patología , Insulinoma/prevención & control , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/prevención & control , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA