Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Immunol ; 212(2): 271-283, 2024 01 15.
Article in English | MEDLINE | ID: mdl-37982696

ABSTRACT

Highly self-reactive T cells are censored from the repertoire by both central and peripheral tolerance mechanisms upon receipt of high-affinity TCR signals. Clonal deletion is considered a major driver of central tolerance; however, other mechanisms such as induction of regulatory T cells and functional impairment have been described. An understanding of the interplay between these different central tolerance mechanisms is still lacking. We previously showed that impaired clonal deletion to a model tissue-restricted Ag did not compromise tolerance. In this study, we determined that murine T cells that failed clonal deletion were rendered functionally impaired in the thymus. Programmed cell death protein 1 (PD-1) was induced in the thymus and was required to establish cell-intrinsic tolerance to tissue-restricted Ag in CD8+ thymocytes independently of clonal deletion. In bone marrow chimeras, tolerance was not observed in PD-L1-deficient recipients, but tolerance was largely maintained following adoptive transfer of tolerant thymocytes or T cells to PD-L1-deficient recipients. However, CRISPR-mediated ablation of PD-1 in tolerant T cells resulted in broken tolerance, suggesting different PD-1 signaling requirements for establishing versus maintaining tolerance. Finally, we showed that chronic exposure to high-affinity Ag supported the long-term maintenance of tolerance. Taken together, our study identifies a critical role for PD-1 in establishing central tolerance in autoreactive T cells that escape clonal deletion. It also sheds light on potential mechanisms of action of anti-PD-1 pathway immune checkpoint blockade and the development of immune-related adverse events.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Mice , Animals , Programmed Cell Death 1 Receptor/genetics , Central Tolerance , CD8-Positive T-Lymphocytes , Thymus Gland , Antigens , Immune Tolerance
2.
J Immunol ; 208(9): 2131-2140, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35396221

ABSTRACT

Several unique waves of γδ T cells are generated solely in the fetal/neonatal thymus, whereas additional γδ T cell subsets are generated in adults. One intriguing feature of γδ T cell development is the coordination of differentiation and acquisition of effector function within the fetal thymus; however, it is less clear whether this paradigm holds true in adult animals. In this study, we investigated the relationship between maturation and thymic export of adult-derived γδ thymocytes in mice. In the Rag2pGFP model, immature (CD24+) γδ thymocytes expressed high levels of GFP whereas only a minority of mature (CD24-) γδ thymocytes were GFP+ Similarly, most peripheral GFP+ γδ T cells were immature. Analysis of γδ recent thymic emigrants (RTEs) indicated that most γδ T cell RTEs were CD24+ and GFP+, and adoptive transfer experiments demonstrated that immature γδ thymocytes can mature outside the thymus. Mature γδ T cells largely did not recirculate to the thymus from the periphery; rather, a population of mature γδ thymocytes that produced IFN-γ or IL-17 remained resident in the thymus for at least 60 d. These data support the existence of two populations of γδ T cell RTEs in adult mice: a majority subset that is immature and matures in the periphery after thymic emigration, and a minority subset that completes maturation within the thymus prior to emigration. Additionally, we identified a heterogeneous population of resident γδ thymocytes of unknown functional importance. Collectively, these data shed light on the generation of the γδ T cell compartment in adult mice.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets , Animals , Emigration and Immigration , Lymphocyte Activation , Mice , Thymocytes
3.
Clin Exp Immunol ; 213(1): 67-75, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37137518

ABSTRACT

The immune system is composed of a variety of different T-cell lineages distributed through both secondary lymphoid tissue and non-lymphoid tissue. The intestinal epithelium is a critical barrier surface that contains numerous intraepithelial lymphocytes that aid in maintaining homeostasis at that barrier. This review focuses on T-cell receptor αß (TCRαß) CD8αα intraepithelial lymphocytes, and how recent advances in the field clarify how this unique T-cell subset is selected, matures, and functions in the intestines. We consider how the available evidence reveals a story of ontogeny starting from agonist selection of T cells in the thymus and finishing through the specific signaling environment of the intestinal epithelium. We conclude with how this story raises further key questions about the development of different ontogenic waves of TCRαß CD8αα IEL and their importance for intestinal epithelial homeostasis.


Subject(s)
CD8-Positive T-Lymphocytes , Intraepithelial Lymphocytes , Animals , Mice , CD8 Antigens , Receptors, Antigen, T-Cell, alpha-beta , Intestines , Intestinal Mucosa , Lymphocytes , Mice, Inbred C57BL
4.
Brain Behav Immun ; 114: 80-93, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37544463

ABSTRACT

Decades of research into chronic pain has deepened our understanding of the cellular mechanisms behind this process. However, a failure to consider the biological variable of sex has limited the application of these breakthroughs into clinical application. In the present study, we investigate fundamental differences in chronic pain between male and female mice resulting from inflammatory activation of the innate immune system. We provide evidence that female mice are more sensitive to the effects of macrophages. Injecting small volumes of media conditioned by either unstimulated macrophages or macrophages stimulated by the inflammatory molecule TNFα lead to increased pain sensitivity only in females. Interestingly, we find that TNFα conditioned media leads to a more rapid resolution of mechanical hypersensitivity and altered immune cell recruitment to sites of injury. Furthermore, male and female macrophages exhibit differential polarization characteristics and motility after TNFα stimulation, as well as a different profile of cytokine secretions. Finally, we find that the X-linked gene Tlr7 is critical in the facilitating the adaptive resolution of pain in models of acute and chronic inflammation in both sexes. Altogether, these findings suggest that although the cellular mechanisms of pain resolution may differ between the sexes, the study of these differences may yield more targeted approaches with clinical applications.

5.
J Immunol ; 205(2): 329-334, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32540993

ABSTRACT

A layer of mucus functions to segregate contents of the intestinal lumen from the intestinal epithelium. The MUC2 mucin is the primary constituent of intestinal mucus and plays critical protective roles against luminal microbes and other noxious agents. In this study, we investigated whether MUC2 helps maintain CD8 T cell tolerance toward intestinal luminal Ags by gavaging wild-type and Muc2-/- mice with a model Ag and monitoring immune responses posttreatment. We report that orally delivered OVA rapidly disseminates through the blood of Muc2-/- (but not control) mice and causes immune activation of Ag-specific CD8 T cells at both local and distal sites. Further, the administration of oral OVA to Muc2-/- mice led to its presentation by thymic dendritic cells and the deletion of Ag-specific thymocytes. Collectively, our findings suggest that intestinal mucus helps limit the shaping of the TCR repertoire of developing thymocytes by intestinal luminal Ags.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Intestines/physiology , Mucin-2/metabolism , Mucus/metabolism , Administration, Oral , Animals , Antigens/immunology , Cell Differentiation , Cell Proliferation , Clonal Deletion , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics
6.
Transfusion ; 61(4): 1247-1257, 2021 04.
Article in English | MEDLINE | ID: mdl-33481275

ABSTRACT

BACKGROUND: Irradiation of red blood cells (RBCs) inactivates residual donor T lymphocytes to prevent transfusion-associated graft-vs-host disease (TA-GVHD) but can have adverse effects on recipients and inventory management. Reported incidence of TA-GVHD is lower when leukoreduced RBCs and older blood products are transfused; therefore, the impact of leukoreduction and storage was evaluated as an alternative prevention strategy. STUDY DESIGN AND METHODS: Effectiveness of leukoreduction filters on white blood cell (WBC) proliferation was evaluated by filtering buffy coat (BC) products and isolating residual WBCs. Additionally, leukoreduced RBCs were spiked with 5 × 106 WBCs on Day 21 of hypothermic storage, then stored and processed on Days 7, 14, and 21 to obtain residual WBCs to investigate the impact of hypothermic storage on their viability and proliferative ability. Viability of residual WBCs was assessed by staining with annexin V and an antibody cocktail for flow cytometry analysis. Proliferative ability was assessed by placing carboxyfluorescein diacetate succinimidyl ester-labeled residual WBCs into culture for 6 days with phytohemagglutinin before flow cytometry assessment. RESULTS: Filtration of BC units depleted WBCs, particularly T lymphocytes, to 0.001% ± 0.003% cells/unit, although proliferative activity remained consistent with prefiltration levels of WBCs. WBCs in stored RBCs remained viable even on Day 21 of storage; however, the proliferative activity decreased to 0.24% ± 0.41%. CONCLUSIONS: Hypothermic storage of RBCs for 21 days or more is sufficient to inactivate T lymphocytes, which may help prevent TA-GVHD when irradiated RBCs are not available.


Subject(s)
Cryobiology/methods , Erythrocytes/physiology , Leukocyte Reduction Procedures/methods , Transfusion Reaction/prevention & control , Blood Preservation/methods , Cell Proliferation/physiology , Cell Proliferation/radiation effects , Erythrocyte Transfusion/adverse effects , Erythrocytes/radiation effects , Filtration , Flow Cytometry/methods , Humans , Incidence , Leukocyte Reduction Procedures/statistics & numerical data , Leukocytes/immunology , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects , Time Factors , Transfusion Reaction/epidemiology , Transfusion Reaction/immunology
7.
J Physiol ; 598(19): 4321-4338, 2020 10.
Article in English | MEDLINE | ID: mdl-32721035

ABSTRACT

KEY POINTS: 25-Hydroxyvitamin D (25OHD) is a partial agonist of TRPV1 whereby 25OHD can weakly activate TRPV1 yet antagonize the stimulatory effects of the full TRPV1 agonists capsaicin and oleoyl dopamine. 25OHD binds to TRPV1 within the same vanilloid binding pocket as capsaicin. 25OHD inhibits the potentiating effects of PKC-mediated TRPV1 activity. 25OHD reduces T-cell activation and trigeminal neuron calcium signalling mediated by TRPV1 activity. These results provide evidence that TRPV1 is a novel receptor for the biological actions of vitamin D in addition to the well-documented effects of vitamin D upon the nuclear vitamin D receptor. The results may have important implications for our current understanding of certain diseases where TRPV1 and vitamin D deficiency have been implicated, such as chronic pain and autoimmune diseases, such as type 1 diabetes. ABSTRACT: The capsaicin receptor TRPV1 plays an important role in nociception, inflammation and immunity and its activity is regulated by exogenous and endogenous lipophilic ligands. As vitamin D is lipophilic and involved in similar biological processes as TRPV1, we hypothesized that it directly regulates TRPV1 activity and function. Our calcium imaging and electrophysiological data demonstrate that vitamin D (25-hydroxyvitamin D (25OHD) and 1,25-hydroxyvitamin D (1,25OHD)) can weakly activate TRPV1 at physiologically relevant concentrations (100 nM). Furthermore, both 25OHD and 1,25OHD can inhibit capsaicin-induced TRPV1 activity (IC50  = 34.3 ± 0.2 and 11.5 ± 0.9 nM, respectively), but not pH-induced TRPV1 activity, suggesting that vitamin D interacts with TRPV1 in the same region as the TRPV1 agonist capsaicin. This hypothesis is supported by our in silico TRPV1 structural modelling studies, which place 25OHD in the same binding region as capsaicin. 25OHD also attenuates PKC-dependent TRPV1 potentiation via interactions with a known PKC phospho-acceptor residue in TRPV1. To provide evidence for a physiological role for the interaction of vitamin D with TRPV1, we employed two different cellular models known to express TRPV1: mouse CD4+ T-cells and trigeminal neurons. Our results indicate that 25OHD reduces TRPV1-induced cytokine release from T-cells and capsaicin-induced calcium activity in trigeminal neurons. In summary, we provide evidence that vitamin D is a novel endogenous regulator of TRPV1 channel activity that may play an important physiological role in addition to its known effects through the canonical nuclear vitamin D receptor pathway.


Subject(s)
Transient Receptor Potential Channels , Animals , Capsaicin/pharmacology , Mice , Neurons , Rats, Sprague-Dawley , TRPV Cation Channels , Vitamin D/pharmacology
8.
J Immunol ; 199(9): 3147-3157, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28947542

ABSTRACT

Negative selection against highly self-reactive thymocytes is critical for preventing autoimmunity. Thymocyte deletion, anergy induction, and agonist selection are all forms of negative selection that can occur following a high-affinity TCR signal. Of Bim and Nur77, two TCR-induced proteins with proapoptotic function, Bim has been shown to be important for clonal deletion in several model systems, whereas Nur77 was often dispensable. However, Nur77 has been reported to influence other aspects of T cell development by mechanisms that may not be related to its proapoptotic function. In this study, we examined the role of Nur77 during thymocyte development in the presence and absence of Bim to separate apoptotic from nonapoptotic functions of Nur77. Polyclonal Bim-/- and Bim-/-Nur77-/- mice exhibited comparable accumulation of high-affinity signaled CD4+CD8+ double-positive thymocytes and CD8+ and CD4+ single-positive thymocytes. However, combined Bim and Nur77 deficiency increased the frequency of thymic Foxp3+ T regulatory cells and Foxp3-FR4hiCD73hi anergic phenotype CD4+ T cells compared with Bim-/- mice, suggesting that Nur77 expression impairs the development of nonconventional tolerance-inducing cell fates. Using the OT-I RIP-mOVA model, we found that Nur77 deficiency did not substantially impact clonal deletion nor did it exacerbate the defect in clonal deletion in the absence of Bim. However, additional loss of Nur77 in the absence of Bim led to diabetes induction, suggesting that Nur77 promotes tolerance in this context. Together, these data reveal novel nondeletional roles for Nur77 that differ between T cell subsets and have implications for self-tolerance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/physiology , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/immunology , Clonal Deletion/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
9.
J Immunol ; 199(3): 965-973, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28659353

ABSTRACT

Neonatal and adult T cells differ in their effector functions. Although it is known that cell-intrinsic differences in mature T cells contribute to this phenomenon, the factors involved remain unclear. Given emerging evidence that the binding strength of a TCR for self-peptide presented by MHC (self-pMHC) impacts T cell function, we sought to determine whether altered thymic selection influences the self-reactivity of the TCR repertoire during ontogeny. We found that conventional and regulatory T cell subsets in the thymus of neonates and young mice expressed higher levels of cell surface CD5, a surrogate marker for TCR avidity for self-pMHC, as compared with their adult counterparts, and this difference in self-reactivity was independent of the germline bias of the neonatal TCR repertoire. The increased binding strength of the TCR repertoire for self-pMHC in neonates was not solely due to reported defects in clonal deletion. Rather, our data suggest that thymic selection is altered in young mice such that thymocytes bearing TCRs with low affinity for self-peptide are not efficiently selected into the neonatal repertoire, and stronger TCR signals accompany both conventional and regulatory T cell selection. Importantly, the distinct levels of T cell self-reactivity reflect physiologically relevant differences based on the preferential expansion of T cells from young mice to fill a lymphopenic environment. Therefore, differences in thymic selection in young versus adult mice skew the TCR repertoire, and the relatively higher self-reactivity of the T cell pool may contribute to the distinct immune responses observed in neonates.


Subject(s)
Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Thymocytes/immunology , Adult , Aging , Animals , Animals, Newborn , CD5 Antigens/genetics , CD5 Antigens/immunology , Cell Differentiation , Clonal Selection, Antigen-Mediated , Fetal Blood , Humans , Infant, Newborn , Lymphocyte Activation , Mice , Protein Binding , Self Tolerance , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology
10.
J Immunol ; 197(5): 1743-53, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27465532

ABSTRACT

T cell development is dependent on the migration of progenitor cells from the bone marrow to the thymus. Upon reaching the thymus, progenitors undergo a complex developmental program that requires inputs from various highly conserved signaling pathways including the Notch and Wnt pathways. To date, Ras signaling has not been implicated in the very earliest stages of T cell differentiation, but members of a family of Ras activators called RasGRPs have been shown to be involved at multiple stages of T cell development. We examined early T cell development in mice lacking RasGRP1, RasGRP3, and RasGRPs 1 and 3. We report that RasGRP1- and RasGRP3-deficient thymi show significantly reduced numbers of early thymic progenitors (ETPs) relative to wild type thymi. Furthermore, RasGRP1/3 double-deficient thymi show significant reductions in ETP numbers compared with either RasGRP1 or RasGRP3 single-deficient thymi, suggesting that both RasGRP1 and RasGRP3 regulate the generation of ETPs. In addition, competitive bone marrow chimera experiments reveal that RasGRP1/3 double-deficient progenitors intrinsically generate ETPs less efficiently than wild type progenitors. Finally, RasGRP1/3-deficient progenitors show impaired migration toward the CCR9 ligand, CCL25, suggesting that RasGRP1 and RasGRP3 may regulate progenitor entry into the thymus through a CCR9-dependent mechanism. These data demonstrate that, in addition to Notch and Wnt, the highly conserved Ras pathway is critical for the earliest stages of T cell development and further highlight the importance of Ras signaling during thymocyte maturation.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Guanine Nucleotide Exchange Factors/metabolism , T-Lymphocytes/physiology , Thymocytes/physiology , Thymus Gland/physiology , ras Guanine Nucleotide Exchange Factors/metabolism , Animals , B-Lymphocytes/immunology , Cell Proliferation , Chemokines, CC/immunology , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Lymphocyte Activation , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/physiology , Mice , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/physiology , Receptors, CCR/immunology , Signal Transduction , T-Lymphocytes/immunology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , ras Guanine Nucleotide Exchange Factors/deficiency , ras Guanine Nucleotide Exchange Factors/genetics
11.
J Immunol ; 194(6): 2643-53, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25687757

ABSTRACT

Negative selection, primarily mediated through clonal deletion of self-reactive thymocytes, is critical for establishing self-tolerance and preventing autoimmunity. Recent studies suggest that the molecular mechanisms of negative selection differ depending on the thymic compartment and developmental stage at which thymocytes are deleted. Using the physiological HY(cd4) TCR transgenic model of negative selection against ubiquitous self-antigen, we previously found that one of the principal mediators implicated in clonal deletion, Bim, is required for caspase-3 activation but is ultimately dispensable for negative selection. On the basis of these data, we hypothesized that Nur77, another molecule thought to be a key mediator of clonal deletion, could be responsible for Bim-independent deletion. Despite comparable Nur77 induction in thymocytes during negative selection, Bim deficiency resulted in an accumulation of high-affinity-signaled thymocytes as well as impairment in caspase-mediated and caspase-independent cell death. Although these data suggested that Bim may be required for Nur77-mediated cell death, we found that transgenic Nur77 expression was sufficient to induce apoptosis independently of Bim. However, transgenic Nur77-induced apoptosis was significantly inhibited in the context of TCR signaling, suggesting that endogenous Nur77 could be similarly regulated during negative selection. Although Nur77 deficiency alone did not alter positive or negative selection, combined deficiency in Bim and Nur77 impaired clonal deletion efficiency and significantly increased positive selection efficiency. Collectively, these data shed light on the different roles for Bim and Nur77 during ubiquitous Ag-mediated clonal deletion and highlight potential differences from their reported roles in tissue-restricted Ag-mediated clonal deletion.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Autoantigens/immunology , Clonal Deletion/immunology , Membrane Proteins/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/immunology , Proto-Oncogene Proteins/immunology , Thymocytes/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/immunology , Caspase 3/metabolism , Cells, Cultured , Clonal Deletion/genetics , Enzyme Activation/immunology , Female , Flow Cytometry , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Thymocytes/cytology , Thymocytes/metabolism
12.
Nat Rev Immunol ; 5(10): 772-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16200080

ABSTRACT

In the past few years, there has been a flurry of discoveries and advancements in our understanding of how the thymus prepares T cells to exist at peace in normal healthy tissue: that is, to be self-tolerant. In the thymus, one of the main mechanisms of T-cell central tolerance is clonal deletion, although the selection of regulatory T cells is also important and is gaining enormous interest. In this Review, we discuss the emerging consensus about which models of clonal deletion are most physiological, and we review recent data that define the molecular mechanisms of central tolerance.


Subject(s)
Clonal Deletion/immunology , Immune Tolerance , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Humans , Thymus Gland/immunology
13.
Proc Natl Acad Sci U S A ; 109(3): 893-8, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22215602

ABSTRACT

Positive and negative selection of thymocytes in the thymus are critical for the development of a mature and self-tolerant T-cell repertoire. The proapoptotic Bcl-2 family member Bim is important for negative selection by inducing apoptosis in thymocytes receiving a strong signal through their antigen receptor. However, in the case of ubiquitous self-antigens (UbA), Bim is not required for the clonal deletion of self-reactive thymocytes, suggesting the existence of nonapoptotic clonal deletion mechanisms. Unlike UbA, clonal deletion to tissue-restricted antigens (TRAs) requires positive selection and CCR7-mediated migration to the medulla. This led us to hypothesize that Bim is required for the latter. To study the role of Bim in clonal deletion to TRA, we constructed bone marrow (BM) chimeras using OT-I Bim-deficient or -sufficient donor bone marrow and recipients that express membrane bound chicken ovalbumin under control of the rat insulin promoter (Rip-mOVA). We found that clonal deletion to TRA was completely abrogated in the absence of Bim and large numbers of mature OT-I CD8 T cells survived in the periphery. Despite the large numbers of autoreactive T cells, the chimeras did not develop diabetes and OT-I Bim-deficient T cells from these chimeras were functionally impaired. Collectively, these data provide unique evidence of a differential, thymocyte-intrinsic, molecular requirement downstream of the T-cell receptor (TCR) for clonal deletion to UbA versus TRA and highlight the profound ability of other tolerance mechanisms to control T-cell autoreactivity in the absence of thymic clonal deletion.


Subject(s)
Antigens/immunology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/immunology , Clonal Deletion/immunology , Membrane Proteins/metabolism , Organ Specificity/immunology , Proto-Oncogene Proteins/metabolism , Thymus Gland/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Autoantigens/immunology , Bcl-2-Like Protein 11 , CD8-Positive T-Lymphocytes/immunology , Chickens , Insulin/genetics , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/deficiency , Rats , Recombinant Fusion Proteins/metabolism
14.
J Biol Chem ; 287(53): 44083-96, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23144462

ABSTRACT

Aortic aneurysm is dilation of the aorta primarily due to degradation of the aortic wall extracellular matrix (ECM). Tissue inhibitors of metalloproteinases (TIMPs) inhibit matrix metalloproteinases (MMPs), the proteases that degrade the ECM. Timp3 is the only ECM-bound Timp, and its levels are altered in the aorta from patients with abdominal aortic aneurysm (AAA). We investigated the causal role of Timp3 in AAA formation. Infusion of angiotensin II (Ang II) using micro-osmotic (Alzet) pumps in Timp3(-/-) male mice, but not in wild type control mice, led to adverse remodeling of the abdominal aorta, reduced collagen and elastin proteins but not mRNA, and elevated proteolytic activities, suggesting excess protein degradation within 2 weeks that led to formation of AAA by 4 weeks. Intriguingly, despite early up-regulation of MMP2 in Timp3(-/-)Ang II aortas, additional deletion of Mmp2 in these mice (Timp3(-/-)/Mmp2(-/-)) resulted in exacerbated AAA, compromised survival due to aortic rupture, and inflammation in the abdominal aorta. Reconstitution of WT bone marrow in Timp3(-/-)/Mmp2(-/-) mice reduced inflammation and prevented AAA in these animals following Ang II infusion. Treatment with a broad spectrum MMP inhibitor (PD166793) prevented the Ang II-induced AAA in Timp3(-/-) and Timp3(-/-)/Mmp2(-/-) mice. Our study demonstrates that the regulatory function of TIMP3 is critical in preventing adverse vascular remodeling and AAA. Hence, replenishing TIMP3, a physiological inhibitor of a number of metalloproteinases, could serve as a therapeutic approach in limiting AAA development or expansion.


Subject(s)
Angiotensin II/metabolism , Aortic Aneurysm, Abdominal/metabolism , Receptors, Virus/deficiency , Angiotensin II/genetics , Animals , Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/genetics , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Virus/genetics
15.
Cell Rep ; 38(11): 110512, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35294874

ABSTRACT

Germinal centers (GCs) are essential for antibody affinity maturation. GC B cells have a unique repertoire of cell surface glycans compared with naive B cells, yet functional roles for changes in glycosylation in the GC have yet to be ascribed. Detection of GCs by the antibody GL7 reflects a downregulation in ligands for CD22, an inhibitory co-receptor of the B cell receptor. To test a functional role for downregulation of CD22 ligands in the GC, we generate a mouse model that maintains CD22 ligands on GC B cells. With this model, we demonstrate that glycan remodeling plays a critical role in the maintenance of B cells in the GC. Sustained expression of CD22 ligands induces higher levels of apoptosis in GC B cells, reduces memory B cell and plasma cell output, and delays affinity maturation of antibodies. These defects are CD22 dependent, demonstrating that downregulation of CD22 ligands on B cells plays a critical function in the GC.


Subject(s)
Germinal Center , Receptors, Antigen, B-Cell , Animals , B-Lymphocytes , Glycosylation , Ligands , Mice , Polysaccharides/metabolism , Receptors, Antigen, B-Cell/metabolism
16.
J Biol Chem ; 285(24): 18928-38, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20400506

ABSTRACT

Calnexin is a molecular chaperone and a component of the quality control of the secretory pathway. We have generated calnexin gene-deficient mice (cnx(-/-)) and showed that calnexin deficiency leads to myelinopathy. Calnexin-deficient mice were viable with no discernible effects on other systems, including immune function, and instead they demonstrated dysmyelination as documented by reduced conductive velocity of nerve fibers and electron microscopy analysis of sciatic nerve and spinal cord. Myelin of the peripheral and central nervous systems of cnx(-/-) mice was disorganized and decompacted. There were no abnormalities in neuronal growth, no loss of neuronal fibers, and no change in fictive locomotor pattern in the absence of calnexin. This work reveals a previously unrecognized and important function of calnexin in myelination and provides new insights into the mechanisms responsible for myelin diseases.


Subject(s)
Calnexin/genetics , Calnexin/physiology , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Animals , Animals, Newborn , Calnexin/metabolism , Cell Membrane/metabolism , Electrophysiology/methods , Endoplasmic Reticulum/metabolism , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Folding , Sciatic Nerve/metabolism , Sciatic Nerve/ultrastructure , Spinal Cord/metabolism , Spinal Cord/ultrastructure
17.
J Exp Med ; 202(1): 111-21, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15998791

ABSTRACT

Sequential rearrangement of the T cell receptor for antigen (TCR) beta and alpha chains is a hallmark of thymocyte development. This temporal control is lost in TCR transgenics because the alpha chain is expressed prematurely at the CD4- CD8- double negative (DN) stage. To test the importance of this, we expressed the HY alpha chain at the physiological CD4+ CD8+ double positive (DP) stage. The reduced DP and increased DN cellularity typically seen in TCR transgenics was not observed when the alpha chain was expressed at the appropriate stage. Surprisingly, antigen-driven selection events were also altered. In male mice, thymocyte deletion now occurred at the single positive or medullary stage. In addition, no expansion of CD8 alpha alpha intestinal intraepithelial lymphocytes (IELs) was observed, despite the fact that HY transgenics have been used to model IEL development. Collectively, these data establish the importance of proper timing of TCR expression in thymic development and selection and emphasize the need to use models that most accurately reflect the physiologic process.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Clonal Deletion , Digestive System/cytology , Digestive System/immunology , Gene Expression , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
18.
Immunol Cell Biol ; 89(1): 16-26, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20956988

ABSTRACT

Clonal selection of T cells occurs in the thymus and is responsible for generating a useful and functional repertoire of T cells. Aberrations in clonal selection result in altered T-cell homeostasis in the secondary lymphoid organs ranging from an absence of T cells to an overabundance of autoreactive T cells. The advent of new technologies facilitating the manipulation of the mouse genome has helped refine our understanding of the molecular and genetic pathways involved in clonal selection and has also revealed a high degree of complexity. Herein, we attempt to review recent advances in thymic selection processes, achieved mostly through genetic manipulations.


Subject(s)
Cell Differentiation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Cell Differentiation/genetics , Clonal Deletion/immunology , Humans , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
19.
J Immunol ; 183(12): 7761-7, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19933852

ABSTRACT

T cell education in the thymus is critical for establishing a functional, yet self-tolerant, T cell repertoire. Negative selection is a key process in enforcing self-tolerance. There are many questions that surround the mechanism of negative selection, but it is currently held that apoptosis initiated by Bim and/or Nur77 is critical for negative selection. Recent studies, however, have questioned the necessity of Bim in maintaining both central and peripheral T cell tolerance. To reconcile these apparently contradictory findings, we examined the role of Bim in negative selection in the well-characterized, physiological HY(cd4) mouse model. We found that while Bim expression was required for CD4(+)CD8(+) double-positive thymocyte apoptosis, it was not required for negative selection. Furthermore, Bim deficiency did not alter the frequency or affinity of male reactive cells that escape negative selection in an oligoclonal repertoire. Collectively, these studies indicate that negative selection occurs efficiently in the absence of apoptosis and suggest that the current paradigm of negative selection requiring apoptosis be revisited.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , Autoantigens/biosynthesis , Cell Differentiation/immunology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Autoantigens/metabolism , Bcl-2-Like Protein 11 , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Clone Cells , Female , H-Y Antigen/immunology , Immune Tolerance/genetics , Ligands , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , T-Lymphocyte Subsets/cytology , Thymus Gland/cytology
20.
J Neuroimmunol ; 354: 577529, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33676084

ABSTRACT

Peripheral nerve injury frequently evokes chronic neuropathic pain. This is initiated by a transient inflammatory response that leads to persistent excitation of dorsal root ganglion (DRG) neurons by inflammatory cytokines such as interleukin 1ß(IL-1ß). In non-neuronal cells such as lymphocytes, interleukin 1 exerts actions at attomolar (aM; 10-18 M) concentrations. We now report that DRG neurons in defined-medium, neuron-enriched culture display increased excitability following 5-6 d exposure of 1aM IL-1ß. This response is mediated in part by type 1 interleukin receptors and involves decreased function of putative KCa1.1 channels. This finding provides new insights into the neuroimmune interactions responsible for neuropathic pain.


Subject(s)
Ganglia, Spinal/metabolism , Interleukin-1beta/metabolism , Neuralgia/metabolism , Sensory Receptor Cells/metabolism , Animals , Ganglia, Spinal/drug effects , Interleukin-1beta/pharmacology , Mice , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL