Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Immunol ; 198(1): 249-256, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27895179

ABSTRACT

Thymic dendritic cells (DC) delete self-antigen-specific thymocytes, and drive development of Foxp3-expressing immunoregulatory T cells. Unlike medullary thymic epithelial cells, which express and present peripheral self-antigen, DC must acquire self-antigen to mediate thymic negative selection. One such mechanism entails the transfer of surface MHC-self peptide complexes from medullary thymic epithelial cells to thymic DC. Despite the importance of thymic DC cross-dressing in negative selection, the factors that regulate the process and the capacity of different thymic DC subsets to acquire MHC and stimulate thymocytes are poorly understood. In this study intercellular MHC transfer by thymic DC subsets was investigated using an MHC-mismatch-based in vitro system. Thymic conventional DC (cDC) subsets signal regulatory protein α (SIRPα+) and CD8α+ readily acquired MHC class I and II from thymic epithelial cells but plasmacytoid DC were less efficient. Intercellular MHC transfer was donor-cell specific; thymic DC readily acquired MHC from TEC plus thymic or splenic DC, whereas thymic or splenic B cells were poor donors. Furthermore DC origin influenced cross-dressing; thymic versus splenic DC exhibited an increased capacity to capture TEC-derived MHC, which correlated with direct expression of EpCAM by DC. Despite similar capacities to acquire MHC-peptide complexes, thymic CD8α+ cDC elicited increased T cell stimulation relative to SIRPα+ cDC. DC cross-dressing was cell-contact dependent and unaffected by lipid raft disruption of donor TEC. Furthermore, blocking PI3K signaling reduced MHC acquisition by thymic CD8α+ cDC and plasmacytoid DC but not SIRPα+ cDC. These findings demonstrate that multiple parameters influence the efficiency of and distinct mechanisms drive intercellular MHC transfer by thymic DC subsets.


Subject(s)
Dendritic Cells/immunology , Major Histocompatibility Complex/immunology , Self Tolerance/immunology , Thymus Gland/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Cell Separation , Epithelial Cells/immunology , Female , Flow Cytometry , Male , Mice , Real-Time Polymerase Chain Reaction
2.
Eur J Immunol ; 46(10): 2352-2362, 2016 10.
Article in English | MEDLINE | ID: mdl-27501268

ABSTRACT

Dysregulation of negative selection contributes to T-cell-mediated autoimmunity, such as type 1 diabetes. The events regulating thymic negative selection, however, are ill defined. Work by our group and others suggest that negative selection is inefficient early in ontogeny and increases with age. This study examines temporal changes in negative selection and the thymic DC compartment. Peptide-induced thymocyte deletion in vivo was reduced in newborn versus 4-week-old NOD mice, despite a similar sensitivity of the respective thymocytes to apoptosis induction. The temporal increase in negative selection corresponded with an elevated capacity of thymic antigen-presenting cells to stimulate T cells, along with altered subset composition and function of resident DC. The frequency of signal regulatory protein α+ (SIRPα+ ) and plasmacytoid DCs was increased concomitant with a decrease in CD8α+ DC in 4-week-old NOD thymi. Importantly, 4-week-old versus newborn thymic SIRPα+ DC exhibited increased antigen processing and presentation via the MHC class II but not class I pathway, coupled with an enhanced T-cell stimulatory capacity not seen in thymic plasmacytoid DC and CD8α+ DC. These findings indicate that the efficiency of thymic DC-mediated negative selection is limited early after birth, and increases with age paralleling expansion of functionally superior thymic SIRPα+ DC.


Subject(s)
Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Receptors, Immunologic/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Animals , Apoptosis , CD8 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Clonal Selection, Antigen-Mediated , Humans , Lymphocyte Activation , Mice , Mice, Inbred NOD
3.
J Immunol ; 191(12): 5858-66, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24198282

ABSTRACT

Inefficient thymic negative selection of self-specific T cells is associated with several autoimmune diseases, including type 1 diabetes. The factors that influence the efficacy of thymic negative selection, as well as the kinetics of thymic output of autoreactive T cells remain ill-defined. We investigated thymic production of ß cell-specific T cells using a thymus-transplantation model. Thymi from different aged NOD mice, representing distinct stages of type 1 diabetes, were implanted into NOD.scid recipients, and the diabetogenicity of the resulting T cell pool was examined. Strikingly, the development of diabetes-inducing ß cell-specific CD4(+) and CD8(+) T cells was regulated in an age-dependent manner. NOD.scid recipients of newborn NOD thymi developed diabetes. However, recipients of thymi from 7- and 10-d-old NOD donor mice remained diabetes-free and exhibited a progressive decline in islet infiltration and ß cell-specific CD4(+) and CD8(+) T cells. A similar temporal decrease in autoimmune infiltration was detected in some, but not all, tissues of recipient mice implanted with thymi from NOD mice lacking expression of the autoimmune regulator transcription factor, which develop multiorgan T cell-mediated autoimmunity. In contrast, recipients of 10 d or older thymi lacked diabetogenic T cells but developed severe colitis marked by increased effector T cells reactive to intestinal microbiota. These results demonstrate that thymic development of autoreactive T cells is limited to a narrow time window and occurs in a reciprocal manner compared with colonic microbiota-responsive T cells in NOD mice.


Subject(s)
Aging/immunology , Autoimmunity/physiology , Clonal Selection, Antigen-Mediated , Diabetes Mellitus, Type 1/immunology , Mice, Inbred NOD/immunology , Thymus Gland/immunology , Adoptive Transfer , Animals , Animals, Newborn , Animals, Suckling , Autoantigens/immunology , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colitis/etiology , Colitis/immunology , Colon/immunology , Colon/microbiology , Colon/pathology , Cytotoxicity, Immunologic , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Female , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphoid Tissue/pathology , Mice , Mice, Knockout , Mice, SCID , Organ Specificity , Pancreas/immunology , Pancreas/pathology , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/pathology , Salivary Glands/immunology , Salivary Glands/pathology , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Thymus Gland/growth & development , Thymus Gland/pathology , Thymus Gland/transplantation , Transcription Factors/deficiency , Transcription Factors/physiology , AIRE Protein
4.
Proc Natl Acad Sci U S A ; 106(12): 4810-5, 2009 Mar 24.
Article in English | MEDLINE | ID: mdl-19251650

ABSTRACT

T cell-mediated autoimmune diseases such as type 1 diabetes (T1D) are believed to be the result in part of inefficient negative selection of self-specific thymocytes. However, the events regulating thymic negative selection are not fully understood. In the current study, we demonstrate that nonobese diabetic (NOD) mice lacking expression of the Mer tyrosine kinase (MerTK) have reduced inflammation of the pancreatic islets and fail to develop diabetes. Furthermore, NOD mice deficient in MerTK expression (Mer(-/-)) exhibit a reduced frequency of beta cell-specific T cells independent of immunoregulatory effectors. The establishment of bone marrow chimeric mice demonstrated that the block in beta cell autoimmunity required hematopoietic-derived cells lacking MerTK expression. Notably, fetal thymic organ cultures and self-peptide administration showed increased thymic negative selection in Mer(-/-) mice. Finally, thymic dendritic cells (DC) prepared from Mer(-/-) mice exhibited an increased capacity to induce thymocyte apoptosis in a peptide-specific manner in vitro. These findings provide evidence for a unique mechanism involving MerTK-mediated regulation of thymocyte negative selection and thymic DC, and suggest a role for MerTK in contributing to beta cell autoimmunity.


Subject(s)
Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Selection, Genetic , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Animals , Bone Marrow Cells/cytology , Dendritic Cells/enzymology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Female , Immunity , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred NOD , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , Thymus Gland/cytology , c-Mer Tyrosine Kinase
5.
JCI Insight ; 6(17)2021 09 08.
Article in English | MEDLINE | ID: mdl-34314385

ABSTRACT

Immunotherapies are needed in the clinic that effectively suppress ß cell autoimmunity and reestablish long-term self-tolerance in type 1 diabetes. We previously demonstrated that nondepleting anti-CD4 (αCD4) and αCD8α antibodies establish rapid and indefinite remission in recent-onset diabetic NOD mice. Diabetes reversal by coreceptor therapy (CoRT) is induced by suppression of pathogenic effector T cells (Teffs) and the selective egress of T cells from the pancreatic lymph nodes and islets that remain free of infiltration in the long term. Here, we defined CoRT-induced events regulating early Teff function and pancreatic residency, and long-term tolerance. TCR-driven gene expression controlling autoreactive Teff expansion and proinflammatory activity was suppressed by CoRT, and islet T cell egress was dependent on sphingosine-1 phosphate. In both murine and human T cells, CoRT upregulated the Foxo1 transcriptional axis, which in turn was required for suppression and efficient pancreatic egress of Teffs. Interestingly, long-term tolerance induced in late-preclinical NOD mice was marked by reseeding of the pancreas by a reduced CD8+ Teff pool exhibiting an exhausted phenotype. Notably, PD-1 blockade, which rescues exhausted Teffs, resulted in diabetes onset in protected animals. These findings demonstrate that CoRT has distinct intrinsic effects on Teffs that impact events early in induction and later in maintenance of self-tolerance.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Immune Tolerance , Immunotherapy/methods , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Mice , Mice, Inbred NOD
6.
Front Immunol ; 11: 624568, 2020.
Article in English | MEDLINE | ID: mdl-33679717

ABSTRACT

Type 1 diabetes (T1D) is widely considered to be a T cell driven autoimmune disease resulting in reduced insulin production due to dysfunction/destruction of pancreatic ß cells. Currently, there continues to be a need for immunotherapies that selectively reestablish persistent ß cell-specific self-tolerance for the prevention and remission of T1D in the clinic. The utilization of monoclonal antibodies (mAb) is one strategy to target specific immune cell populations inducing autoimmune-driven pathology. Several mAb have proven to be clinically safe and exhibit varying degrees of efficacy in modulating autoimmunity, including T1D. Traditionally, mAb therapies have been used to deplete a targeted cell population regardless of antigenic specificity. However, this treatment strategy can prove detrimental resulting in the loss of acquired protective immunity. Nondepleting mAb have also been applied to modulate the function of immune effector cells. Recent studies have begun to define novel mechanisms associated with mAb-based immunotherapy that alter the function of targeted effector cell pools. These results suggest short course mAb therapies may have persistent effects for regaining and maintaining self-tolerance. Furthermore, the flexibility to manipulate mAb properties permits the development of novel strategies to target multiple antigens and/or deliver therapeutic drugs by a single mAb molecule. Here, we discuss current and potential future therapeutic mAb treatment strategies for T1D, and T cell-mediated autoimmunity.


Subject(s)
Antibodies, Monoclonal , Diabetes Mellitus, Type 1 , Immunotherapy , Insulin-Secreting Cells , Lymphocyte Depletion , T-Lymphocytes , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Humans , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
7.
Front Immunol ; 11: 615371, 2020.
Article in English | MEDLINE | ID: mdl-33603744

ABSTRACT

T cell receptor (TCR) signaling influences multiple aspects of CD4+ and CD8+ T cell immunobiology including thymic development, peripheral homeostasis, effector subset differentiation/function, and memory formation. Additional T cell signaling cues triggered by co-stimulatory molecules and cytokines also affect TCR signaling duration, as well as accessory pathways that further shape a T cell response. Type 1 diabetes (T1D) is a T cell-driven autoimmune disease targeting the insulin producing ß cells in the pancreas. Evidence indicates that dysregulated TCR signaling events in T1D impact the efficacy of central and peripheral tolerance-inducing mechanisms. In this review, we will discuss how the strength and nature of TCR signaling events influence the development of self-reactive T cells and drive the progression of T1D through effects on T cell gene expression, lineage commitment, and maintenance of pathogenic anti-self T cell effector function.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Models, Immunological , Receptors, Antigen, T-Cell/immunology , T-Lymphocyte Subsets/immunology , Autoantigens/immunology , Autoimmunity/immunology , B7-H1 Antigen/immunology , Cell Lineage , Clonal Selection, Antigen-Mediated , Cytokines/immunology , DNA Methylation , Gene Expression Regulation/immunology , Histone Code , Homeostasis , Humans , Immunologic Memory/immunology , Lymphocyte Activation , Programmed Cell Death 1 Receptor/immunology , Self Tolerance/immunology , Signal Transduction/immunology , Thymus Gland/immunology
8.
Front Immunol ; 9: 1891, 2018.
Article in English | MEDLINE | ID: mdl-30166987

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that is generally considered to be T cell-driven. Accordingly, most strategies of immunotherapy for T1D prevention and treatment in the clinic have targeted the T cell compartment. To date, however, immunotherapy has had only limited clinical success. Although certain immunotherapies have promoted a protective effect, efficacy is often short-term and acquired immunity may be impacted. This has led to the consideration of combining different approaches with the goal of achieving a synergistic therapeutic response. In this review, we will discuss the status of various T1D therapeutic strategies tested in the clinic, as well as possible combinatorial approaches to restore ß cell tolerance.


Subject(s)
Autoimmunity/drug effects , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Immunomodulation/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/immunology , Animals , Cytokines/metabolism , Cytokines/therapeutic use , Diabetes Mellitus, Type 1/metabolism , Disease Susceptibility , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Immunotherapy/adverse effects , Immunotherapy/methods , Insulin-Secreting Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Adv Healthc Mater ; 7(18): e1800341, 2018 09.
Article in English | MEDLINE | ID: mdl-30051618

ABSTRACT

Type 1 diabetes (T1D) is a common autoimmune disease with no cure. T1D subjects are dependent on daily exogenous insulin administration, due to the loss of functional insulin-producing ß cells. Needed are immunotherapies that prevent and/or treat T1D. One approach of immunotherapy is to administer an autoantigen to selectively tolerize diabetogenic effector T cells without global immunosuppression. To date, however, strategies of antigen-specific immunotherapy are largely ineffective in the clinic. Using an antigen-specific approach, a biodegradable polymeric delivery vehicle, acetalated dextran microparticles (Ace-DEX MPs), is applied and T1D development is prevented through coadministration of the immunosuppressant rapamycin and the diabetogenic peptide P31 (Rapa/P31/MPs), via alterations of both innate and adaptive immunity. Ex vivo, adoptively transferred CD4+ T cells exhibit reduced proliferation and an increased ratio of FoxP3+ to IFNγ+ T cells. In vitro analysis indicates dendritic cells exhibit a less mature phenotype following coculture with Rapa/P31/MPs, which results in reduced CD4+ T cell proliferation and proinflammatory cytokine production (IFNγ and IL-2), but promotes PD-1 expression. Together these results demonstrate Ace-DEX MP-based antigen-specific therapy effectively tolerizes diabetogenic CD4+ T cells to prevent T1D, thereby demonstrating one of the first successful attempts of T1D prevention using a single-formulation particulate delivery platform.


Subject(s)
Dextrans/chemistry , Diabetes Mellitus, Type 1/prevention & control , Pancreatic Polypeptide/chemistry , Sirolimus/chemistry , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred BALB C , Pancreatic Polypeptide/administration & dosage , Sirolimus/administration & dosage , Sirolimus/therapeutic use
10.
Front Immunol ; 8: 1898, 2017.
Article in English | MEDLINE | ID: mdl-29312356

ABSTRACT

Inflammation is typically induced in response to a microbial infection. The release of proinflammatory cytokines enhances the stimulatory capacity of antigen-presenting cells, as well as recruits adaptive and innate immune effectors to the site of infection. Once the microbe is cleared, inflammation is resolved by various mechanisms to avoid unnecessary tissue damage. Autoimmunity arises when aberrant immune responses target self-tissues causing inflammation. In type 1 diabetes (T1D), T cells attack the insulin producing ß cells in the pancreatic islets. Genetic and environmental factors increase T1D risk by in part altering central and peripheral tolerance inducing events. This results in the development and expansion of ß cell-specific effector T cells (Teff) which mediate islet inflammation. Unlike protective immunity where inflammation is terminated, autoimmunity is sustained by chronic inflammation. In this review, we will highlight the key events which initiate and sustain T cell-driven pancreatic islet inflammation in nonobese diabetic mice and in human T1D. Specifically, we will discuss: (i) dysregulation of thymic selection events, (ii) the role of intrinsic and extrinsic factors that enhance the expansion and pathogenicity of Teff, (iii) defects which impair homeostasis and suppressor activity of FoxP3-expressing regulatory T cells, and (iv) properties of ß cells which contribute to islet inflammation.

11.
Arch Immunol Ther Exp (Warsz) ; 58(6): 449-57, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20872284

ABSTRACT

Events ongoing in the thymus are critical for deleting developing thymocytes specific for tissue antigens, and establishing self-tolerance within the T cell compartment. Aberrant thymic negative selection, however, is believed to generate a repertoire with increased self-reactivity, which in turn can contribute to the development of T cell-mediated autoimmunity. In this review, mechanisms that regulate the efficacy of negative selection and influence the deletion of autoreactive thymocytes will be discussed.


Subject(s)
Autoimmunity , Clonal Deletion , Immune Tolerance , Lymphocyte Activation , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Humans
12.
J Immunol ; 180(9): 5784-8, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424695

ABSTRACT

CTL that possess a high functional avidity are known to be optimal for the clearance of pathogens in vivo. We have shown that the amount of peptide encountered by a CD8+ CTL determines its functional avidity. Notably, in these studies nonprofessional APC were used. However, it is mature dendritic cells (DC) that are predominantly responsible for the activation of naive T cells in vivo. Whether DC also direct dose dependent-differences in avidity is unknown. In this work we examined the ability of mature DC presenting a high vs low level of peptide to generate CTL of distinct avidities. In contrast to what was observed with nonprofessional APC, CTL generated by stimulation with mature DC were of high avidity regardless of the amount of peptide presented. This DC property may promote generation of highly effective CTL that retain plasticity, which would allow the tuning of avidity in the periphery to promote optimal pathogen recognition and clearance.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Dendritic Cells/cytology , Dose-Response Relationship, Immunologic , Lymphocyte Activation/drug effects , Mice , Peptides/pharmacology , T-Lymphocytes, Cytotoxic/cytology
13.
J Immunol ; 179(2): 748-51, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617563

ABSTRACT

The property of functional avidity is recognized to be of critical importance in determining pathogen clearance. An unresolved question with regard to this property is whether distinct naive subsets exist that display inherent differences in their peptide sensitivity requirements for activation, i.e., functional avidity, or whether differences in peptide sensitivity are induced following peptide encounter. In this study, we demonstrate that naive populations that can give rise to both high- and low-avidity cells do not contain subsets that exhibit differences in the amount of peptide required for activation. Furthermore, we show that an individual T cell clone can generate both high- and low-avidity effectors. The work presented here provides the first formal demonstration that an individual cell can give rise to both high- and low-avidity progeny, suggesting that avidity modulation at the level of an individual cell may play an important role in the CD8(+) T cell response generated in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Animals , Clone Cells , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology
14.
Immunology ; 122(2): 167-78, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17484768

ABSTRACT

The generation of an optimal CD8(+) cytotoxic T lymphocyte (CTL) response is critical for the clearance of many intracellular pathogens. Previous studies suggest that one contributor to an optimal immune response is the presence of CD8(+) cells exhibiting high functional avidity. In this regard, CD8 expression has been shown to contribute to peptide sensitivity. Here, we investigated the ability of naive splenocytes to modulate CD8 expression according to the concentration of stimulatory peptide antigen. Our results showed that the level of CD8 expressed was inversely correlated with the amount of peptide used for the primary stimulation, with higher concentrations of antigen resulting in lower expression of both CD8alpha and CD8beta. Importantly the ensuing CD8(low) and CD8(high) CTL populations were not the result of the selective outgrowth of naive CD8(+) T-cell subpopulations expressing distinct levels of CD8. Subsequent encounter with peptide antigen resulted in continued modulation of both the absolute level and the isoform of CD8 expressed and in the functional avidity of the responding cells. We propose that CD8 cell surface expression is not a static property, but can be modulated to 'fine tune' the sensitivity of responding CTL to a defined concentration of antigen.


Subject(s)
Antibody Affinity , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Differentiation/immunology , Cells, Cultured , Dose-Response Relationship, Immunologic , Gene Expression Regulation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/immunology , RNA, Messenger/genetics , Spleen/immunology
15.
Cell Immunol ; 227(2): 148-55, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15135297

ABSTRACT

CD28 has been shown to play an important role in T cell activation. Among the downstream events associated with CD28 engagement is the reorganization of the cytoskeleton resulting in lipid raft aggregation. In our previous studies we investigated the involvement of lipid rafts in the activation of high avidity CD8+ T lymphocytes, which recognize cells bearing very low levels of peptide antigen, versus low avidity cells, which require high levels of peptide antigen. In these studies we found that high avidity cells were much more sensitive to lipid raft disruption compared to low avidity cells. Given the important role for CD28 in lipid raft reorganization and our previous finding that high avidity cells are extremely dependent on lipid raft integrity, we hypothesized that high avidity cells could not be generated in the absence of CD28. Surprisingly, we have found that the absence of CD28 does not alter the ability to generate high or low avidity CD8+ T cells. In fact high and low avidity lines generated in parallel from CD28-deficient and WT mice exhibited very similar requirements for peptide antigen. We next compared the effect of lipid raft disruption on the activation of high versus low avidity cells from CD28-deficient and WT mice. While high avidity cells generated from WT mice exhibited the expected dependence on lipid raft integrity, high avidity cells from CD28-deficient mice were not affected. These data suggest that the lines generated from the CD28-deficient mice have developed alternative strategies to promote high sensitivity to peptide antigen.


Subject(s)
CD28 Antigens/physiology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Membrane Microdomains/physiology , Animals , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/physiology
SELECTION OF CITATIONS
SEARCH DETAIL