Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Dev Biol ; 511: 53-62, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38593904

ABSTRACT

Early embryonic development is a finely orchestrated process that requires precise regulation of gene expression coordinated with morphogenetic events. TATA-box binding protein-associated factors (TAFs), integral components of transcription initiation coactivators like TFIID and SAGA, play a crucial role in this intricate process. Here we show that disruptions in TAF5, TAF12 and TAF13 individually lead to embryonic lethality in the mouse, resulting in overlapping yet distinct phenotypes. Taf5 and Taf12 mutant embryos exhibited a failure to implant post-blastocyst formation, and Taf5 mutants have aberrant lineage specification within the inner cell mass. In contrast, Taf13 mutant embryos successfully implant and form egg-cylinder stages but fail to initiate gastrulation. Strikingly, we observed a depletion of pluripotency factors in TAF13-deficient embryos, including OCT4, NANOG and SOX2, highlighting an indispensable role of TAF13 in maintaining pluripotency. Transcriptomic analysis revealed distinct gene targets affected by the loss of TAF5, TAF12 and TAF13. Thus, we propose that TAF5, TAF12 and TAF13 convey locus specificity to the TFIID complex throughout the mouse genome.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , TATA-Binding Protein Associated Factors , Animals , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/genetics , Mice , Embryonic Development/genetics , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Female , Blastocyst/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Gastrulation/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Embryo, Mammalian/metabolism
2.
Nat Commun ; 14(1): 5585, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37696797

ABSTRACT

The success of the CD8 T cell-mediated immune response against infections and tumors depends on the formation of a long-lived memory pool, and the protection of effector cells from exhaustion. The advent of checkpoint blockade therapy has significantly improved anti-tumor therapeutic outcomes by reversing CD8 T cell exhaustion, but fails to generate effector cells with memory potential. Here, using in vivo mouse models, we show that let-7 miRNAs determine CD8 T cell fate, where maintenance of let-7 expression during early cell activation results in memory CD8 T cell formation and tumor clearance. Conversely, let-7-deficiency promotes the generation of a terminal effector population that becomes vulnerable to exhaustion and cell death in immunosuppressive environments and fails to reject tumors. Mechanistically, let-7 restrains metabolic changes that occur during T cell activation through the inhibition of the PI3K/AKT/mTOR signaling pathway and production of reactive oxygen species, potent drivers of terminal differentiation and exhaustion. Thus, our results reveal a role for let-7 in the time-sensitive support of memory formation and the protection of effector cells from exhaustion. Overall, our data suggest a strategy in developing next-generation immunotherapies by preserving the multipotency of effector cells rather than enhancing the efficacy of differentiation.


Subject(s)
CD8-Positive T-Lymphocytes , MicroRNAs , Phosphatidylinositol 3-Kinases , Animals , Mice , Antibodies , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Neoplasms , Phosphatidylinositol 3-Kinases/genetics , MicroRNAs/genetics , MicroRNAs/metabolism
3.
Mol Immunol ; 157: 129-141, 2023 05.
Article in English | MEDLINE | ID: mdl-37018939

ABSTRACT

Following activation, CD4 T cells undergo metabolic and transcriptional changes as they respond to external cues and differentiate into T helper (Th) cells. T cells exhibit plasticity between Th phenotypes in highly inflammatory environments, such as colitis, in which high levels of IL-6 promote plasticity between regulatory T (Treg) cells and Th17 cells. Protein Kinase C theta (PKCθ) is a T cell-specific serine/threonine kinase that promotes Th17 differentiation while negatively regulating Treg differentiation. Liver kinase B1 (LKB1), also a serine/threonine kinase and encoded by Stk11, is necessary for Treg survival and function. Stk11 can be alternatively spliced to produce a short variant (Stk11S) by transcribing a cryptic exon. However, the contribution of Stk11 splice variants to Th cell differentiation has not been previously explored. Here we show that in Th17 cells, the heterogeneous ribonucleoprotein, hnRNPLL, mediates Stk11 splicing into its short splice variant, and that Stk11S expression is diminished when Hnrnpll is depleted using siRNA knock-down approaches. We further show that PKCθ regulates hnRNPLL and, thus, Stk11S expression in Th17 cells. We provide additional evidence that exposing induced (i)Tregs to IL-6 culminates in Stk11 splicing downstream of PKCθAltogether our data reveal a yet undescribed outside-in signaling pathway initiated by IL-6, that acts through PKCθ and hnRNPLL to regulate Stk11 splice variants and facilitate Th17 cell differentiation. Furthermore, we show for the first time, that this pathway can also be initiated in developing iTregs exposed to IL-6, providing mechanistic insight into iTreg phenotypic stability and iTreg to Th17 cell plasticity.


Subject(s)
Cell Plasticity , Interleukin-6 , Protein Kinase C-theta/metabolism , Interleukin-6/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , CD4-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Cell Differentiation , Protein Isoforms/metabolism , Th17 Cells/metabolism
4.
Front Immunol ; 14: 1272918, 2023.
Article in English | MEDLINE | ID: mdl-38179041

ABSTRACT

The phenomenon of intercellular transfer of cellular material, including membranes, cytoplasm, and even organelles, has been observed for decades. The functional impact and molecular mechanisms of such transfer in the immune system remain largely elusive due to the absence of a robust in vivo model. Here, we introduce a new tumor mouse model, where tumor cells express the soluble ultra-bright fluorescent protein ZsGreen, which allows detection and measurement of intercellular transfer of cytoplasm from tumor cells to infiltrating immune cells. We found that in addition to various types of myeloid lineage cells, a large fraction of T regulatory cells and effector CD8 T cells acquire tumor material. Based on the distribution of tumor-derived ZsGreen, the majority of T cells integrate captured cytoplasm into their own, while most myeloid cells store tumor material in granules. Furthermore, scRNA-seq analysis revealed significant alterations in transcriptomes of T cells that acquired tumor cell cytoplasm, suggesting potential impact on T cell function. We identified that the participation of T cells in intercellular transfer requires cell-cell contact and is strictly dependent on the activation status of T lymphocytes. Finally, we propose to name the described phenomenon of intercellular transfer for tumor infiltrating T cells the "mosquito effect".


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocytes, Tumor-Infiltrating , Mice , Animals , Cytoplasm , Cytosol , Disease Models, Animal
5.
Mol Immunol ; 120: 67-73, 2020 04.
Article in English | MEDLINE | ID: mdl-32085976

ABSTRACT

CD8 T cells are among the most vigorous soldiers of the immune system that fight viral infections and cancer. CD8 T cell development, maintenance, activation and differentiation are under the tight control of multiple transcriptional and post-transcriptional networks. Over the last two decades it has become clear that non-coding RNAs (ncRNAs), which consist of microRNAs (miRNAs) and long ncRNAs (lncRNAs), have emerged as global biological regulators. While our understanding of the function of specific miRNAs has increased since the discovery of RNA interference, it is still very limited, and the field of lncRNAs is just starting to blossom. Here we will summarize our knowledge on the role of ncRNAs in CD8 T cell biology, including differentiation into memory and exhausted cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , RNA, Untranslated/genetics , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation , Humans , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , MicroRNAs/genetics , MicroRNAs/immunology , RNA Interference , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , RNA, Untranslated/immunology
6.
Front Immunol ; 10: 955, 2019.
Article in English | MEDLINE | ID: mdl-31130952

ABSTRACT

Maintaining the diversity and constant numbers of naïve T cells throughout the organism's lifetime is necessary for efficient immune responses. Naïve T cell homeostasis, which consists of prolonged survival, occasional proliferation and enforcement of quiescence, is tightly regulated by multiple signaling pathways which are in turn controlled by various transcription factors. However, full understanding of the molecular mechanisms underlying the maintenance of the peripheral T cell pool has not been achieved. In the present study, we demonstrate that T cell-specific deficiency in let-7 miRNAs results in peripheral T cell lymphopenia resembling that of Dicer1 knockout mice. Deletion of let-7 leads to profound T cell apoptosis while overexpression prevents it. We further show that in the absence of let-7, T cells cannot sustain optimal levels of the pro-survival factor Bcl2 in spite of the intact IL-7 signaling, and re-expression of Bcl2 in let-7 deficient T cells completely rescues the survival defect. Thus, we have uncovered a novel let-7-dependent mechanism of post-transcriptional regulation of naïve T cell survival in vivo.


Subject(s)
MicroRNAs/immunology , T-Lymphocytes/immunology , Animals , DEAD-box RNA Helicases/genetics , Lymph Nodes/cytology , Mice, Knockout , MicroRNAs/genetics , Ribonuclease III/genetics
7.
Front Immunol ; 10: 3125, 2019.
Article in English | MEDLINE | ID: mdl-32010153

ABSTRACT

Multiple sclerosis (MS) is a disabling demyelinating autoimmune disorder of the central nervous system (CNS) which is driven by IL-23- and IL-1ß-induced autoreactive Th17 cells that traffic to the CNS and secrete proinflammatory cytokines. Th17 pathogenicity in MS has been correlated with the dysregulation of microRNA (miRNA) expression, and specific miRNAs have been shown to promote the pathogenic Th17 phenotype. In the present study, we demonstrate, using the animal model of MS, experimental autoimmune encephalomyelitis (EAE), that let-7 miRNAs confer protection against EAE by negatively regulating the proliferation, differentiation and chemokine-mediated migration of pathogenic Th17 cells to the CNS. Specifically, we found that let-7 miRNAs may directly target the cytokine receptors Il1r1 and Il23r, as well as the chemokine receptors Ccr2 and Ccr5. Therefore, our results identify a novel regulatory role for let-7 miRNAs in pathogenic Th17 differentiation during EAE development, suggesting a promising therapeutic application for disease treatment.


Subject(s)
Disease Susceptibility , MicroRNAs/genetics , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Gene Expression Regulation , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Multiple Sclerosis/pathology , RNA Interference , Th17 Cells/cytology
9.
Elife ; 62017 07 24.
Article in English | MEDLINE | ID: mdl-28737488

ABSTRACT

The differentiation of naive CD8 T cells into effector cytotoxic T lymphocytes upon antigen stimulation is necessary for successful antiviral, and antitumor immune responses. Here, using a mouse model, we describe a dual role for the let-7 microRNAs in the regulation of CD8 T cell responses, where maintenance of the naive phenotype in CD8 T cells requires high levels of let-7 expression, while generation of cytotoxic T lymphocytes depends upon T cell receptor-mediated let-7 downregulation. Decrease of let-7 expression in activated T cells enhances clonal expansion and the acquisition of effector function through derepression of the let-7 targets, including Myc and Eomesodermin. Ultimately, we have identified a novel let-7-mediated mechanism, which acts as a molecular brake controlling the magnitude of CD8 T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Cell Differentiation , MicroRNAs/metabolism , Animals , Mice
10.
Nat Immunol ; 16(5): 517-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25848867

ABSTRACT

Lethal-7 (let-7) microRNAs (miRNAs) are the most abundant miRNAs in the genome, but their role in developing thymocytes is unclear. We found that let-7 miRNAs targeted Zbtb16 mRNA, which encodes the lineage-specific transcription factor PLZF, to post-transcriptionally regulate PLZF expression and thereby the effector functions of natural killer T cells (NKT cells). Dynamic upregulation of let-7 miRNAs during the development of NKT thymocytes downregulated PLZF expression and directed their terminal differentiation into interferon-γ (IFN-γ)-producing NKT1 cells. Without upregulation of let-7 miRNAs, NKT thymocytes maintained high PLZF expression and terminally differentiated into interleukin 4 (IL-4)-producing NKT2 cells or IL-17-producing NKT17 cells. Upregulation of let-7 miRNAs in developing NKT thymocytes was signaled by IL-15, vitamin D and retinoic acid. Such targeting of a lineage-specific transcription factor by miRNA represents a previously unknown level of developmental regulation in the thymus.


Subject(s)
Cytokines/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Natural Killer T-Cells/physiology , Thymocytes/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cytotoxicity, Immunologic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Promyelocytic Leukemia Zinc Finger Protein , Protein Binding , RNA Processing, Post-Transcriptional , Tretinoin/metabolism , Up-Regulation , Vitamin D/metabolism
11.
Sci Signal ; 7(340): ra83, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25161318

ABSTRACT

T cell receptor (TCR)-mediated inhibition of interleukin-7 (IL-7) signaling is important for lineage fate determination in the thymus and for T cell survival in the periphery because uninterrupted IL-7 signaling results in T cell death. The initial event in IL-7 signaling is the transactivation of Janus kinases 1 and 3 (Jak1 and Jak3), which are associated with the cytosolic tails of the IL-7 receptor α chain (IL-7Rα) and the γc subunit, the two cell surface proteins that constitute IL-7R. We found that Jak1 is a highly unstable protein with a half-life of only 1.5 hours, so that continuous Jak1 protein synthesis is required to maintain Jak1 protein in sufficient abundance to support IL-7 signaling. However, we also found that Jak1 protein synthesis was acutely reduced by TCR-responsive microRNAs in the miR-17 family, which targeted Jak1 mRNA (messenger RNA) to inhibit its translation. Thus, this study identifies a molecular mechanism by which TCR engagement acutely disrupts IL-7 signaling.


Subject(s)
Gene Expression Regulation/immunology , Janus Kinase 1/genetics , MicroRNAs/metabolism , RNA, Messenger/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-7/metabolism , Signal Transduction/immunology , Animals , Blotting, Western , Flow Cytometry , Immunoprecipitation , Janus Kinase 1/biosynthesis , Luciferases , Mice , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-7/antagonists & inhibitors
12.
Cell ; 154(6): 1326-41, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034254

ABSTRACT

Thymic selection requires signaling by the protein tyrosine kinase Lck to generate T cells expressing αß T cell antigen receptors (TCR). For reasons not understood, the thymus selects only αßTCR that are restricted by major histocompatibility complex (MHC)-encoded determinants. Here, we report that Lck proteins that were coreceptor associated promoted thymic selection of conventionally MHC-restricted TCR, but Lck proteins that were coreceptor free promoted thymic selection of MHC-independent TCR. Transgenic TCR with MHC-independent specificity for CD155 utilized coreceptor-free Lck to signal thymic selection in the absence of MHC, unlike any transgenic TCR previously described. Thus, the thymus can select either MHC-restricted or MHC-independent αßTCR depending on whether Lck is coreceptor associated or coreceptor free. We conclude that the intracellular state of Lck determines the specificity of thymic selection and that Lck association with coreceptor proteins during thymic selection is the mechanism by which MHC restriction is imposed on a randomly generated αßTCR repertoire.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , T-Lymphocytes/cytology , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , Major Histocompatibility Complex , Mice , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Virus , Signal Transduction , T-Lymphocytes/metabolism , Thymus Gland/immunology
13.
Nat Immunol ; 14(2): 143-51, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23242416

ABSTRACT

The maintenance of naive CD8(+) T cells is necessary for lifelong immunocompetence but for unknown reasons requires signaling via both interleukin 7 (IL-7) and the T cell antigen receptor (TCR). We now report that naive CD8(+) T cells required IL-7 signaling to be intermittent, not continuous, because prolonged IL-7 signaling induced naive CD8(+) T cells to proliferate, produce interferon-γ (IFN-γ) and undergo IFN-γ-triggered cell death. Homeostatic engagement of the TCR interrupted IL-7 signaling and thereby supported the survival and quiescence of CD8(+) T cells. However, CD8(+) T cells with insufficient TCR affinity for self ligands received prolonged IL-7 signaling and died during homeostasis. In this study we identified regulation of the duration of IL-7 signaling by homeostatic engagement of the TCR as the basis for in vivo CD8(+) T cell homeostasis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Interleukin-7/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Death/immunology , Cell Proliferation , Cell Survival/immunology , Gene Expression Regulation , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-7/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Time Factors
14.
Nat Immunol ; 13(6): 569-78, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544394

ABSTRACT

Clonal deletion of autoreactive thymocytes is important for self-tolerance, but the intrathymic signals that induce clonal deletion have not been clearly identified. We now report that clonal deletion during negative selection required CD28-mediated costimulation of autoreactive thymocytes at the CD4(+)CD8(lo) intermediate stage of differentiation. Autoreactive thymocytes were prevented from undergoing clonal deletion by either a lack of CD28 costimulation or transgenic overexpression of the antiapoptotic factors Bcl-2 or Mcl-1, with surviving thymocytes differentiating into anergic CD4(-)CD8(-) double-negative thymocytes positive for the T cell antigen receptor αß subtype (TCRαß) that 'preferentially' migrated to the intestine, where they re-expressed CD8α and were sequestered as CD8αα(+) intraepithelial lymphocytes (IELs). Our study identifies costimulation by CD28 as the intrathymic signal required for clonal deletion and identifies CD8αα(+) IELs as the developmental fate of autoreactive thymocytes that survive negative selection.


Subject(s)
Cell Differentiation/immunology , Clonal Deletion/immunology , Receptors, Antigen, T-Cell/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , CD28 Antigens/immunology , CD4 Antigens/immunology , CD8 Antigens/immunology , Flow Cytometry , Immune Tolerance/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Thymocytes/cytology , Thymus Gland/cytology
15.
Immunity ; 36(1): 79-91, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22209676

ABSTRACT

Major histocompatibility complex (MHC) restriction is the cardinal feature of T cell antigen recognition and is thought to be intrinsic to αß T cell receptor (TCR) structure because of germline-encoded residues that impose MHC specificity. Here, we analyzed αßTCRs from T cells that had not undergone MHC-specific thymic selection. Instead of recognizing peptide-MHC complexes, the two αßTCRs studied here resembled antibodies in recognizing glycosylation-dependent conformational epitopes on a native self-protein, CD155, and they did so with high affinity independently of MHC molecules. Ligand recognition was via the αßTCR combining site and involved the identical germline-encoded residues that have been thought to uniquely impose MHC specificity, demonstrating that these residues do not only promote MHC binding. This study demonstrates that, without MHC-specific thymic selection, αßTCRs can resemble antibodies in recognizing conformational epitopes on MHC-independent ligands.


Subject(s)
Antibody Specificity , Epitopes, T-Lymphocyte/metabolism , Major Histocompatibility Complex , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Animals , Gene Deletion , Ligands , Mice , Molecular Sequence Data , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Virus/metabolism , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
16.
J Exp Med ; 202(1): 73-84, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15998789

ABSTRACT

MHC molecules associated with autoimmunity possess known structural features that limit the repertoire of peptides that they can present. Such limitation gives a selective advantage to TCRs that rely on interaction with the MHC itself, rather than with the peptide residues. At the same time, negative selection is impaired because of the lack of negatively selecting peptide ligands. The combination of these factors may predispose to autoimmunity. We found that mice with an MHC class II-peptide repertoire reduced to a single complex demonstrated various autoimmune reactions. Transgenic mice bearing a TCR (MM14.4) cloned from such a mouse developed severe autoimmune dermatitis. Although MM14.4 originated from a CD4+ T cell, dermatitis was mediated by CD8+ T cells. It was established that MM14.4+ is a highly promiscuous TCR with dual MHC class I/MHC class II restriction. Furthermore, mice with a limited MHC-peptide repertoire selected elevated numbers of TCRs with dual MHC class I/MHC class II restriction, a likely source of autoreactivity. Our findings may help to explain the link between MHC class I responses that are involved in major autoimmune diseases and the well-established genetic linkage of these diseases with MHC class II.


Subject(s)
Autoimmunity , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dermatitis/etiology , Dermatitis/immunology , Dermatitis/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...