Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38352301

ABSTRACT

Oncogene activity rewires cellular transcription, creating new transcription networks to which cancer cells become addicted, by mechanisms that are still poorly understood. Using human and mouse models of T cell acute lymphoblastic leukemia (T-ALL), we identify an essential nuclear role for CHMP5, a cytoplasmic endosomal sorting complex required for transport (ESCRT) protein, in establishing and maintaining the T-ALL transcriptional program. Nuclear CHMP5 promoted the T-ALL gene program by augmenting recruitment of the co-activator BRD4 by the histone acetyl transferase p300 selectively at enhancers and super-enhancers, an interaction that potentiated H3K27 acetylation at these regulatory enhancers. Consequently, loss of CHMP5 diminished BRD4 occupancy at enhancers and super-enhancers and impaired RNA polymerase II pause release, which resulted in downregulation of key T-ALL genes, notably MYC. Reinforcing its importance in T-ALL pathogenesis, CHMP5 deficiency mitigated chemoresistance in human T-ALL cells and abrogated T-ALL induction by oncogenic NOTCH1 in vivo. Thus, the ESCRT protein CHMP5 is an essential positive regulator of the transcriptional machinery promoting T-ALL disease.

2.
J Clin Invest ; 134(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38226624

ABSTRACT

The suppressor of lin-12-like-HMG-CoA reductase degradation 1 (SEL1L-HRD1) complex of the endoplasmic reticulum-associated degradation (ERAD) machinery is a key cellular proteostasis pathway. Although previous studies have shown ERAD as promoting the development and maintenance of many cell types in mice, its importance to human physiology remained undetermined. In two articles in this issue of the JCI, Qi and colleagues describe four biallelic hypomorphic SEL1L and HRD1 variants that were associated with neurodevelopment disorders, locomotor dysfunction, impaired immunity, and premature death in patients. These pathogenic SEL1L-HRD1 variants shine a light on the critical importance of ERAD in humans and pave the way for future studies dissecting ERAD mechanisms in specific cell types.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Proteins , Ubiquitin-Protein Ligases , Animals , Humans , Mice , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Proteins/genetics , Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Theranostics ; 12(6): 2894-2907, 2022.
Article in English | MEDLINE | ID: mdl-35401837

ABSTRACT

The endoplasmic reticulum unfolded protein response (UPR) is a conserved adaptive signaling in ER homeostasis and has emerged as critical in highly proliferating cells and potential treatment target for acute T-cell lymphoblastic leukemia (T-ALL). Methods: in this study, we assessed the transcriptomic and phenotypic alterations in UPR response of the bone marrow endothelial cells (ECs) in mice engrafted with T-ALL and in bone marrow specimens from patients who have T-ALL. We used PERK inhibitor and generated endothelial specific PERK knockout mice to study the impact of PERK on leukemia progression and hematopoiesis. We performed chromatin immunoprecipitation (ChIP) to study the mechanistic regulation of JAG1 by ATF4. We characterized small extracellular vesicles (SEV) from leukemia-developing mice and studied the effect of SEVs on EC function. Results: we found that T-ALL development induced a robust activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK)-dominant UPR in the bone marrow endothelial vascular niche. The activation of PERK-eIF2a-ATF4 axis remodels the vascular niche, upregulates angiogenic factors including VEGFα and ATF4-regulated JAG1, and suppresses the expression of SCF and CXCL12, which are important to HSC maintenance and regeneration. Further, targeting endothelial PERK significantly improved T-ALL outcome. EC-specific deletion of PERK abolished the aberrant JAG1 up-regulation, improved HSC maintenance, promoted leukemia apoptosis, and improved overall survival. Finally, we showed that small extracellular vesicles are critical mediators of endothelial PERK-eIF2a-ATF4 activation and JAG1 up-regulation in leukemia. Corroborating animal model studies, activation of PERK-ATF4-JAG1 is prominent in human T-ALL bone marrow and T-ALL xenografts. Conclusion: our studies thus revealed for the first time that the leukemia-initiated PERK-ATF4-JAG1 axis plays a critical role in the remodeling of the bone marrow vascular niche and that targeting vascular niche UPR is a potential therapeutic opportunity in T-ALL.


Subject(s)
Endothelial Cells , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Unfolded Protein Response , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Bone Marrow/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Endothelial Cells/metabolism , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Jagged-1 Protein/pharmacology , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , eIF-2 Kinase/metabolism
4.
Elife ; 102021 07 09.
Article in English | MEDLINE | ID: mdl-34240701

ABSTRACT

Signals from the pre-T cell receptor and Notch coordinately instruct ß-selection of CD4-CD8-double negative (DN) thymocytes to generate αß T cells in the thymus. However, how these signals ensure a high-fidelity proteome and safeguard the clonal diversification of the pre-selection TCR repertoire given the considerable translational activity imposed by ß-selection is largely unknown. Here, we identify the endoplasmic reticulum (ER)-associated degradation (ERAD) machinery as a critical proteostasis checkpoint during ß-selection. Expression of the SEL1L-HRD1 complex, the most conserved branch of ERAD, is directly regulated by the transcriptional activity of the Notch intracellular domain. Deletion of Sel1l impaired DN3 to DN4 thymocyte transition and severely impaired mouse αß T cell development. Mechanistically, Sel1l deficiency induced unresolved ER stress that triggered thymocyte apoptosis through the PERK pathway. Accordingly, genetically inactivating PERK rescued T cell development from Sel1l-deficient thymocytes. In contrast, IRE1α/XBP1 pathway was induced as a compensatory adaptation to alleviate Sel1l-deficiency-induced ER stress. Dual loss of Sel1l and Xbp1 markedly exacerbated the thymic defect. Our study reveals a critical developmental signal controlled proteostasis mechanism that enforces T cell development to ensure a healthy adaptive immunity.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/drug effects , Receptors, Notch/metabolism , Thymocytes/metabolism , Animals , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Female , Inflammation , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Proteostasis , Ubiquitin-Protein Ligases/metabolism , X-Box Binding Protein 1/metabolism
5.
Nat Cell Biol ; 22(10): 1162-1169, 2020 10.
Article in English | MEDLINE | ID: mdl-32958856

ABSTRACT

Stem cells need to be protected from genotoxic and proteotoxic stress to maintain a healthy pool throughout life1-3. Little is known about the proteostasis mechanism that safeguards stem cells. Here we report endoplasmic reticulum-associated degradation (ERAD) as a protein quality checkpoint that controls the haematopoietic stem cell (HSC)-niche interaction and determines the fate of HSCs. The SEL1L-HRD1 complex, the most conserved branch of ERAD4, is highly expressed in HSCs. Deletion of Sel1l led to niche displacement of HSCs and a complete loss of HSC identity, and allowed highly efficient donor-HSC engraftment without irradiation. Mechanistic studies identified MPL, the master regulator of HSC identity5, as a bona fide ERAD substrate that became aggregated in the endoplasmic reticulum following ERAD deficiency. Restoration of MPL signalling with an agonist partially rescued the number and reconstitution capacity of Sel1l-deficient HSCs. Our study defines ERAD as an essential proteostasis mechanism to safeguard a healthy stem cell pool by regulating the stem cell-niche interaction.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Hematopoietic Stem Cells/cytology , Intracellular Signaling Peptides and Proteins/physiology , Receptors, Thrombopoietin/metabolism , Stem Cell Niche , Ubiquitin-Protein Ligases/metabolism , Animals , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Thrombopoietin/genetics , Ubiquitin-Protein Ligases/genetics
6.
Methods Mol Biol ; 1998: 219-226, 2019.
Article in English | MEDLINE | ID: mdl-31250305

ABSTRACT

The charged multivesicular body protein-5 (CHMP5) is a member of the endosomal-sorting complex required for transport (ESCRT) that controls membrane-scission events in eukaryotic cells. Recent studies have revealed novel functions of CHMP5 beyond its role in the ESCRT machinery, notably as a critical nonenzymatic regulator of the ubiquitination and subsequent degradation of proteins in immune cells. Here we describe an immunoprecipitation and western blot methodology for assessing CHMP5 activity on client protein ubiquitination in T lymphocytes.


Subject(s)
Blotting, Western/methods , Endosomal Sorting Complexes Required for Transport/metabolism , Immunoprecipitation/methods , Ubiquitination , Endosomes/metabolism , HEK293 Cells , Humans , Jurkat Cells , T-Lymphocytes
7.
Nat Immunol ; 18(7): 780-790, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28553951

ABSTRACT

The acquisition of a protective vertebrate immune system hinges on the efficient generation of a diverse but self-tolerant repertoire of T cells by the thymus through mechanisms that remain incompletely resolved. Here we identified the endosomal-sorting-complex-required-for-transport (ESCRT) protein CHMP5, known to be required for the formation of multivesicular bodies, as a key sensor of thresholds for signaling via the T cell antigen receptor (TCR) that was essential for T cell development. CHMP5 enabled positive selection by promoting post-selection thymocyte survival in part through stabilization of the pro-survival protein Bcl-2. Accordingly, loss of CHMP5 in thymocyte precursor cells abolished T cell development, a phenotype that was 'rescued' by genetic deletion of the pro-apoptotic protein Bim or transgenic expression of Bcl-2. Mechanistically, positive selection resulted in the stabilization of CHMP5 by inducing its interaction with the deubiquitinase USP8. Our results thus identify CHMP5 as an essential component of the post-translational machinery required for T cell development.


Subject(s)
Cell Differentiation/immunology , Endosomal Sorting Complexes Required for Transport/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Animals , Bcl-2-Like Protein 11/immunology , Endopeptidases/immunology , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymocytes/cytology , Ubiquitin Thiolesterase/immunology
8.
Nat Immunol ; 16(8): 829-37, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26147683

ABSTRACT

The transcription factor XBP1 has been linked to the development of highly secretory tissues such as plasma cells and Paneth cells, yet its function in granulocyte maturation has remained unknown. Here we discovered an unexpectedly selective and absolute requirement for XBP1 in eosinophil differentiation without an effect on the survival of basophils or neutrophils. Progenitors of myeloid cells and eosinophils selectively activated the endoribonuclease IRE1α and spliced Xbp1 mRNA without inducing parallel endoplasmic reticulum (ER) stress signaling pathways. Without XBP1, nascent eosinophils exhibited massive defects in the post-translational maturation of key granule proteins required for survival, and these unresolvable structural defects fed back to suppress critical aspects of the transcriptional developmental program. Hence, we present evidence that granulocyte subsets can be distinguished by their differential reliance on secretory-pathway homeostasis.


Subject(s)
Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Eosinophils/immunology , Gene Expression/immunology , Transcription Factors/immunology , Animals , Cell Differentiation/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/genetics , Endoribonucleases/immunology , Endoribonucleases/metabolism , Eosinophils/metabolism , Eosinophils/ultrastructure , Flow Cytometry , Gene Expression Profiling , Granulocyte Precursor Cells/immunology , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/ultrastructure , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , X-Box Binding Protein 1
9.
Nat Commun ; 6: 7562, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26108174

ABSTRACT

Initial events after exposure determine HIV-1 disease progression, underscoring a critical need to understand host mechanisms that interfere with initial viral replication. Although associated with chronic HIV-1 control, it is not known whether interleukin-21 (IL-21) contributes to early HIV-1 immunity. Here we take advantage of tractable primary human lymphoid organ aggregate cultures to show that IL-21 directly suppresses HIV-1 replication, and identify microRNA-29 (miR-29) as an antiviral factor induced by IL-21 in CD4 T cells. IL-21 promotes transcription of all miR-29 species through STAT3, whose binding to putative regulatory regions within the MIR29 gene is enriched by IL-21 signalling. Notably, exogenous IL-21 limits early HIV-1 infection in humanized mice, and lower viremia in vivo is associated with higher miR-29 expression. Together, these findings reveal a novel antiviral IL-21-miR-29 axis that promotes CD4 T-cell-intrinsic resistance to HIV-1 infection, and suggest a role for IL-21 in initial HIV-1 control in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV Infections/metabolism , HIV-1/physiology , Interleukins/pharmacology , MicroRNAs/metabolism , Animals , Antibodies, Monoclonal, Humanized , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Spleen/cytology , Tissue Culture Techniques
10.
Nature ; 508(7494): 103-107, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24670641

ABSTRACT

Cancer cells induce a set of adaptive response pathways to survive in the face of stressors due to inadequate vascularization. One such adaptive pathway is the unfolded protein (UPR) or endoplasmic reticulum (ER) stress response mediated in part by the ER-localized transmembrane sensor IRE1 (ref. 2) and its substrate XBP1 (ref. 3). Previous studies report UPR activation in various human tumours, but the role of XBP1 in cancer progression in mammary epithelial cells is largely unknown. Triple-negative breast cancer (TNBC)--a form of breast cancer in which tumour cells do not express the genes for oestrogen receptor, progesterone receptor and HER2 (also called ERBB2 or NEU)--is a highly aggressive malignancy with limited treatment options. Here we report that XBP1 is activated in TNBC and has a pivotal role in the tumorigenicity and progression of this human breast cancer subtype. In breast cancer cell line models, depletion of XBP1 inhibited tumour growth and tumour relapse and reduced the CD44(high)CD24(low) population. Hypoxia-inducing factor 1α (HIF1α) is known to be hyperactivated in TNBCs. Genome-wide mapping of the XBP1 transcriptional regulatory network revealed that XBP1 drives TNBC tumorigenicity by assembling a transcriptional complex with HIF1α that regulates the expression of HIF1α targets via the recruitment of RNA polymerase II. Analysis of independent cohorts of patients with TNBC revealed a specific XBP1 gene expression signature that was highly correlated with HIF1α and hypoxia-driven signatures and that strongly associated with poor prognosis. Our findings reveal a key function for the XBP1 branch of the UPR in TNBC and indicate that targeting this pathway may offer alternative treatment strategies for this aggressive subtype of breast cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , CD24 Antigen/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Gene Silencing , Humans , Hyaluronan Receptors/metabolism , Mice , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Prognosis , RNA Polymerase II/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/genetics , Unfolded Protein Response , X-Box Binding Protein 1
11.
Nat Immunol ; 13(10): 972-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22961053

ABSTRACT

The transcription factor Foxp3 participates dominantly in the specification and function of Foxp3(+)CD4(+) regulatory T cells (T(reg) cells) but is neither strictly necessary nor sufficient to determine the characteristic T(reg) cell signature. Here we used computational network inference and experimental testing to assess the contribution of other transcription factors to this. Enforced expression of Helios or Xbp1 elicited distinct signatures, but Eos, IRF4, Satb1, Lef1 and GATA-1 elicited exactly the same outcome, acting in synergy with Foxp3 to activate expression of most of the T(reg) cell signature, including key transcription factors, and enhancing occupancy by Foxp3 at its genomic targets. Conversely, the T(reg) cell signature was robust after inactivation of any single cofactor. A redundant genetic switch thus 'locked in' the T(reg) cell phenotype, a model that would account for several aspects of T(reg) cell physiology, differentiation and stability.


Subject(s)
T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , CD4 Antigens/biosynthesis , Cell Differentiation , Computational Biology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Forkhead Transcription Factors/genetics , GATA1 Transcription Factor/genetics , Humans , Ikaros Transcription Factor/biosynthesis , Ikaros Transcription Factor/genetics , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Lymphoid Enhancer-Binding Factor 1/genetics , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Regulatory Factor X Transcription Factors , Serine Endopeptidases/genetics , T-Lymphocytes, Regulatory/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics , X-Box Binding Protein 1
13.
EMBO J ; 31(2): 366-77, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22036949

ABSTRACT

Immature thymocytes are bipotential cells that are signalled during positive selection to become either helper- or cytotoxic-lineage T cells. By tracking expression of lineage determining transcription factors during positive selection, we now report that the Cd8 coreceptor gene locus co-opts any coreceptor protein encoded within it to induce thymocytes to express the cytotoxic-lineage factor Runx3 and to adopt the cytotoxic-lineage fate, findings we refer to as 'coreceptor gene imprinting'. Specifically, encoding CD4 proteins in the endogenous Cd8 gene locus caused major histocompatibility complex class II-specific thymocytes to express Runx3 during positive selection and to differentiate into CD4(+) cytotoxic-lineage T cells. Our findings further indicate that coreceptor gene imprinting derives from the dynamic regulation of specific cis Cd8 gene enhancer elements by positive selection signals in the thymus. Thus, for coreceptor-dependent thymocytes, lineage fate is determined by Cd4 and Cd8 coreceptor gene loci and not by the specificity of T-cell antigen receptor/coreceptor signalling. This study identifies coreceptor gene imprinting as a critical determinant of lineage fate determination in the thymus.


Subject(s)
CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , Lymphopoiesis/genetics , T-Lymphocyte Subsets/cytology , Thymocytes/cytology , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , Cell Lineage/genetics , Clonal Selection, Antigen-Mediated , Core Binding Factor Alpha 3 Subunit/biosynthesis , Core Binding Factor Alpha 3 Subunit/genetics , Cytokines/physiology , DNA, Recombinant/genetics , Enhancer Elements, Genetic/genetics , Gene Knock-In Techniques , Genes, Synthetic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
Nat Immunol ; 11(3): 257-64, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20118929

ABSTRACT

Immature CD4(+)CD8(+) (double-positive (DP)) thymocytes are signaled via T cell antigen receptors (TCRs) to undergo positive selection and become responsive to intrathymic cytokines such as interleukin 7 (IL-7). We report here that cytokine signaling is required for positively selected thymocytes to express the transcription factor Runx3, specify CD8 lineage choice and differentiate into cytotoxic-lineage T cells. In DP thymocytes genetically engineered to be cytokine responsive, IL-7 signaling induced TCR-unsignaled DP thymocytes to express Runx3 and to differentiate into mature CD8(+) T cells, completely circumventing positive selection. We conclude that TCR-mediated positive selection converts DP cells into cytokine-responsive thymocytes, but it is subsequent signaling by intrathymic cytokines that specifies CD8 lineage choice and promotes differentiation into cytotoxic-lineage T cells.


Subject(s)
Cytokines/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Count , Cell Differentiation/immunology , Cell Lineage , Core Binding Factor Alpha 3 Subunit/immunology , Flow Cytometry , Interleukin-7/immunology , Mice , Mice, Knockout , Mice, Transgenic , STAT5 Transcription Factor/immunology , Signal Transduction
15.
Immunity ; 31(3): 480-90, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19747858

ABSTRACT

The lineage fate of developing thymocytes is determined by the persistence or cessation of T cell receptor (TCR) signaling during positive selection, with persistent TCR signaling required for CD4 lineage choice. We show here that transcriptional upregulation of CD4 expression is essential for error-free lineage choice during major histocompatibility complex class II (MHC II)-specific positive selection and is critical for error-free lineage choice in TCR-transgenic mice whose thymocytes compete for the identical selecting ligand. CD4 upregulation occurred for endogenously encoded CD4 coreceptors, but CD4 transgenes were downregulated during positive selection, disrupting MHC II-specific TCR signaling and causing lineage errors regardless of the absolute number or signaling strength of transgenic CD4 proteins. Thus, the kinetics of CD4 coreceptor expression during MHC II-specific positive selection determines the integrity of CD4 lineage choice, revealing an elegant symmetry between coreceptor kinetics and lineage choice.


Subject(s)
CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Lineage , Histocompatibility Antigens Class II/immunology , Up-Regulation , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
16.
J Immunol ; 181(10): 6975-83, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18981117

ABSTRACT

The mechanism by which CD4/CD8 lineage choice is coordinated with TCR specificity during positive selection remains an unresolved problem in immunology. The stochastic/selection model proposes that CD4/CD8 lineage choice in TCR-signaled CD4(+)CD8(+) thymocytes occurs randomly and therefore is highly error-prone. This perspective is strongly supported by "coreceptor rescue" experiments in which transgenic CD4 coreceptors were ectopically expressed on thymocytes throughout their development and caused significant numbers of cells bearing MHC-II-specific TCR to differentiate into mature, CD8 lineage T cells. However, it is not known if forced coreceptor expression actually rescued positively selected thymocytes making an incorrect lineage choice or if it influenced developing thymocytes into making an incorrect lineage choice. We have now reassessed coreceptor rescue and the concept that lineage choice is highly error-prone with a novel CD4 transgene (referred to as E8(I)-CD4) that targets expression of transgenic CD4 coreceptors specifically to thymocytes that have already undergone positive selection and adopted a CD8 lineage fate. Unlike previous CD4 transgenes, the E8(I)-CD4 transgene has no effect on early thymocyte development and cannot itself influence CD4/CD8 lineage choice. We report that the E8(I)-CD4 transgene did in fact induce expression of functional CD4 coreceptor proteins on newly arising CD8 lineage thymocytes precisely at the point in thymic development that transgenic CD4 coreceptors would putatively rescue MHC-II-specific thymocytes that incorrectly adopted the CD8 lineage. However, the E8(I)-CD4 transgene did not reveal any MHC-II-selected thymocytes that adopted the CD8 lineage fate. These results demonstrate that CD4/CD8 lineage choice is neither error-prone nor stochastic.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Major Histocompatibility Complex/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
17.
Nat Rev Immunol ; 8(10): 788-801, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18802443

ABSTRACT

Following successful gene rearrangement at alphabeta T-cell receptor (TCR) loci, developing thymocytes express both CD4 and CD8 co-receptors and undergo a life-or-death selection event, which is known as positive selection, to identify cells that express TCRs with potentially useful ligand specificities. Positively selected thymocytes must then differentiate into either CD4(+) helper T cells or CD8(+) cytotoxic T cells, a crucial decision known as CD4/CD8-lineage choice. In this Review, we summarize recent advances in our understanding of the cellular and molecular events involved in lineage-fate decision and discuss them in the context of the major models of CD4/CD8-lineage choice.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Lineage , Cytokines/immunology , Cytokines/metabolism , Humans , Models, Biological , Protein Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/metabolism
18.
Immunity ; 27(5): 735-50, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18023370

ABSTRACT

The thymus generates major histocompatibility complex (MHC)-restricted alphabetaT cells that only recognize antigenic ligands in association with MHC or MHC-like molecules. We hypothesized that MHC specificity might be imposed on a broader alphabetaTCR repertoire during thymic selection by CD4 and CD8 coreceptors that bind and effectively sequester the tyrosine kinase Lck, thereby preventing T cell receptor (TCR) signaling by non-MHC ligands that do not engage either coreceptor. This hypothesis predicts that, in coreceptor-deficient mice, alphabeta thymocytes would be signaled by non-MHC ligands to differentiate into alphabetaT cells lacking MHC specificity. We now report that MHC-independent alphabetaT cells were indeed generated in mice deficient in both coreceptors as well as MHC ("quad-deficient" mice) and that such mice contained a diverse alphabetaT cell repertoire whose MHC independence was confirmed at the clonal level. We conclude that CD4 and CD8 coreceptors impose MHC specificity on a broader alphabetaTCR repertoire during thymic selection by preventing thymocytes from being signaled by non-MHC ligands.


Subject(s)
CD4 Antigens/immunology , CD8 Antigens/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Blotting, Northern , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation/immunology , Flow Cytometry , Fluorescent Antibody Technique , Immunoprecipitation , Major Histocompatibility Complex/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology
19.
Nat Immunol ; 8(10): 1049-59, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873878

ABSTRACT

T cell immunity requires the long-term survival of T cells that are capable of recognizing self antigens but are not overtly autoreactive. How this balance is achieved remains incompletely understood. Here we identify a homeostatic mechanism that transcriptionally tailors CD8 coreceptor expression in individual CD8+ T cells to the self-specificity of their clonotypic T cell receptor (TCR). 'Coreceptor tuning' results from interplay between cytokine and TCR signals, such that signals from interleukin 7 and other common gamma-chain cytokines transcriptionally increase CD8 expression and thereby promote TCR engagement of self ligands, whereas TCR signals impair common gamma-chain cytokine signaling and thereby decrease CD8 expression. This dynamic interplay induces individual CD8+ T cells to express CD8 in quantities appropriate for the self-specificity of their TCR, promoting the engagement of self ligands, yet avoiding autoreactivity.


Subject(s)
CD8 Antigens/genetics , Interleukin-7/pharmacology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Cytokines/pharmacology , Enhancer Elements, Genetic , Homeostasis , Humans , Mice , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...