Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Nature ; 609(7926): 354-360, 2022 09.
Article in English | MEDLINE | ID: mdl-35978192

ABSTRACT

CD8+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2-is maintained by precursors of exhausted T (TPEX) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6. Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-myb , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunotherapy , L-Selectin/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Viruses/immunology
2.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636132

ABSTRACT

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Subject(s)
Amino Acids/metabolism , CD8-Positive T-Lymphocytes/cytology , Immunologic Memory , Signal Transduction , Amino Acid Transport Systems/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Cycle , Cell Differentiation , Disease Models, Animal , Female , Gene Knock-In Techniques , Lymphocytic Choriomeningitis/immunology , Male , Mice , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology
3.
Eur J Immunol ; 51(5): 1080-1088, 2021 05.
Article in English | MEDLINE | ID: mdl-33521937

ABSTRACT

TCRαß+ CD8α+ CD8ß- intestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely ß2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+ . However, selective depletion of ß2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.


Subject(s)
Clonal Selection, Antigen-Mediated , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Lymphopoiesis , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/cytology , Animals , Biomarkers , Cell Differentiation , Genes, MHC Class I , Immunophenotyping , Intraepithelial Lymphocytes/cytology , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Precursor Cells, T-Lymphoid/cytology , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
4.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33479171

ABSTRACT

Runt domain-related (Runx) transcription factors are essential for early T cell development in mice from uncommitted to committed stages. Single and double Runx knockouts via Cas9 show that target genes responding to Runx activity are not solely controlled by the dominant factor, Runx1. Instead, Runx1 and Runx3 are coexpressed in single cells; bind to highly overlapping genomic sites; and have redundant, collaborative functions regulating genes pivotal for T cell development. Despite stable combined expression levels across pro-T cell development, Runx1 and Runx3 preferentially activate and repress genes that change expression dynamically during lineage commitment, mostly activating T-lineage genes and repressing multipotent progenitor genes. Furthermore, most Runx target genes are sensitive to Runx perturbation only at one stage and often respond to Runx more for expression transitions than for maintenance. Contributing to this highly stage-dependent gene regulation function, Runx1 and Runx3 extensively shift their binding sites during commitment. Functionally distinct Runx occupancy sites associated with stage-specific activation or repression are also distinguished by different patterns of partner factor cobinding. Finally, Runx occupancies change coordinately at numerous clustered sites around positively or negatively regulated targets during commitment. This multisite binding behavior may contribute to a developmental "ratchet" mechanism making commitment irreversible.


Subject(s)
Cell Lineage/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes/immunology , Transcriptome , Animals , Cell Differentiation , Cell Lineage/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Core Binding Factor Alpha 3 Subunit/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Male , Mice , Precursor Cells, T-Lymphoid/cytology , Primary Cell Culture , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Lymphocytes/classification , T-Lymphocytes/cytology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
5.
Exp Hematol ; 95: 1-12, 2021 03.
Article in English | MEDLINE | ID: mdl-33454362

ABSTRACT

T-Cell development is a major branch of lymphoid development and a key output of hematopoiesis, especially in early life, but the molecular requirements for T-cell potential have remained obscure. Considerable advances have now been made toward solving this problem through single-cell transcriptome studies, interfaced with in vitro differentiation assays that monitor potential efficiently at the single-cell level. This review focuses on a series of recent reports studying mouse and human early T-cell precursors, both in the developing fetus and in stringently purified postnatal samples of intrathymic and prethymic T-lineage precursors. Cross-comparison of results reveals a robustly conserved core program in mouse and human, but with some informative and provocative variations between species and between ontogenic states. Repeated findings are the multipotent progenitor regulatory signature of thymus-seeding cells and the proximity of the T-cell program to dendritic cell programs, especially to plasmacytoid dendritic cells in humans.


Subject(s)
Hematopoiesis , Precursor Cells, T-Lymphoid/cytology , Single-Cell Analysis/methods , Animals , Antigens, Differentiation, T-Lymphocyte/analysis , Cell Lineage , Cell Movement , Cell Separation , Cells, Cultured , Dendritic Cells/cytology , Fetus/cytology , Fetus/immunology , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Humans , Mice , Multipotent Stem Cells/cytology , Precursor Cells, T-Lymphoid/classification , Receptors, Antigen, T-Cell, alpha-beta/genetics , Repressor Proteins/physiology , Species Specificity , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/growth & development , Transcriptome , Tumor Suppressor Proteins/physiology
7.
Mediators Inflamm ; 2020: 8867760, 2020.
Article in English | MEDLINE | ID: mdl-33424437

ABSTRACT

BACKGROUND: Infections are an important cause of morbidity and mortality for acute lymphoblastic leukemia (ALL). However, the reports regarding risk factors of induction-related infection are roughly unknown/limited in adult T-ALL during induction chemotherapy. METHODS: We performed a retrospective cohort study for the prevalence and risk predictors of induction-related infection among consecutive T-ALL patients (N = 97) enrolled in a PDT-ALL-LBL clinical trial. Of 97 patients with T-ALL enrolled in the trial, 46 were early T-cell precursor (ETP) ALL and 51 were non-ETP ALL. RESULTS: When compared with non-ETP, ETP ALL subtype was characterized with lower neutrophil count (1.35 × 109/L vs. 8.7 × 109/L, P < 0.001) and lower myeloid percentage in the bone marrow (13.35% vs. 35.31%, P = 0.007). Additionally, ETP ALL had longer neutropenia before diagnosis (P < 0.001), as well as during induction chemotherapy (P < 0.001). Notably, the ETP cohort experienced higher cumulative incidence of clinically documented infections (CDI; 33.33%, P = 0.001), microbiologically documented infections (MDI; 45.24%, P = 0.006), resistant infection (11.9%, P = 0.013), and mixed infection (21.43%, P = 0.003), respectively, than those of the non-ETP cohort. Furthermore, multivariable analysis revealed that T-ALL mixed infection was more likely related to chemotherapy response (OR, 0.025; 95% CI 0.127-0.64; P = 0.012) and identified myeloid percentage as a predictor associated with ETP-ALL mixed infection (OR, 0.915; 95% CI 0.843-0.993; P = 0.033), with ROC-defined cut-off value of 2.24% in ETP cohorts. CONCLUSIONS: Our data for the first time demonstrated that ETP-ALL characterized with impaired myelopoiesis were more susceptible to induction-related infection among T-ALL populations.


Subject(s)
Induction Chemotherapy/adverse effects , Infections/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cells, T-Lymphoid/cytology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Adolescent , Adult , Antineoplastic Agents/pharmacology , Female , Humans , Immunophenotyping , Infections/etiology , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Prevalence , Prognosis , Remission Induction , Retrospective Studies , Risk , Risk Factors , Young Adult
8.
Blood Adv ; 3(20): 2934-2948, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31648315

ABSTRACT

Broader clinical application of umbilical cord blood (UCB), as a source of hematopoietic stem/progenitor cells (HSPCs), is limited by low CD34+ and T-cell numbers, contributing to slow lymphohematopoietic recovery, infection, and relapse. Studies have evaluated the safety, feasibility, and expedited neutrophil recovery associated with the transplantation of CD34+ HSPCs from ex vivo expansion cultures using the aryl hydrocarbon receptor antagonist StemRegenin-1 (SR1). In a phase 1/2 study of 17 patients who received combined SR1-expanded and unexpanded UCB units, a considerable advantage for enhancing T-cell chimerism was not observed. We previously showed that progenitor T (proT) cells generated in vitro from HSPCs accelerated T-cell reconstitution and restored immunity after hematopoietic stem cell transplantation (HSCT). To expedite immune recovery, we hypothesized that SR1-expanded HSPCs together with proT cells could overcome the known T-cell immune deficiency that occurs post-HSCT. Here, we show that SR1-expanded UCB can induce >250-fold expansion of CD34+ HSPCs, which can generate large numbers of proT cells upon in vitro differentiation. When compared with nonexpanded naive proT cells, SR1 proT cells also showed effective thymus-seeding and peripheral T-cell functional capabilities in vivo despite having an altered phenotype. In a competitive transfer approach, both naive and SR1 proT cells showed comparable thymus-engrafting capacities. Single-cell RNA sequencing of peripheral CD3+ T cells from mice injected with either naive or SR1 proT cells revealed functional subsets of T cells with polyclonal T-cell receptor repertoires. Our findings support the use of SR1-expanded UCB grafts combined with proT-cell generation for decreasing T-cell immunodeficiency post-HSCT.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Purines/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement , Cells, Cultured , Clonal Evolution , Coculture Techniques , Fetal Blood/cytology , Gene Expression Profiling , Hematopoietic Stem Cell Transplantation , Humans , Immunophenotyping , Mice , Mice, Transgenic
9.
Immunity ; 51(5): 930-948.e6, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31604687

ABSTRACT

Generation of the first T lymphocytes in the human embryo involves the emergence, migration, and thymus seeding of lymphoid progenitors together with concomitant thymus organogenesis, which is the initial step to establish the entire adaptive immune system. However, the cellular and molecular programs regulating this process remain unclear. We constructed a single-cell transcriptional landscape of human early T lymphopoiesis by using cells from multiple hemogenic and hematopoietic sites spanning embryonic and fetal stages. Among heterogenous early thymic progenitors, one subtype shared common features with a subset of lymphoid progenitors in fetal liver that are known as thymus-seeding progenitors. Unbiased bioinformatics analysis identified a distinct type of pre-thymic lymphoid progenitors in the aorta-gonad-mesonephros (AGM) region. In parallel, we investigated thymic epithelial cell development and potential cell-cell interactions during thymus organogenesis. Together, our data provide insights into human early T lymphopoiesis that prospectively direct T lymphocyte regeneration, which might lead to development of clinical applications.


Subject(s)
Cell Differentiation/genetics , Lymphopoiesis/genetics , Organogenesis/genetics , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/embryology , Biomarkers , Cell Differentiation/immunology , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Lymphopoiesis/immunology , Signal Detection, Psychological , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Transcriptome
10.
Zhonghua Xue Ye Xue Za Zhi ; 40(7): 561-567, 2019 Jul 14.
Article in Chinese | MEDLINE | ID: mdl-32397018

ABSTRACT

Objective: To evaluate the clinical characteristics of T-cell acute leukemia/lymphoma (T-ALL) and explore the prognosis significance of early T-cell precursor leukemia/lymphoma. Methods: A cohort of 126 patients diagnosed with T-ALL from 2008 to 2014 in West China Hospital, Sichuan University were enrolled in this study. They were further categorized by immunophenotype according to the expression of T-cell lineage markers CD1a, CD8, CD5 and one or more stem cell or myeloid markers. The laboratory indicators and prognosis factors were also statistically analyzed. Results: Of all patients, the ratio of male to female was 2.5∶1, with the median age of 25 years old (range 14 to 77) . The percentage of ETP-ALL was up to 47.6%. T-ALL patients showed higher ratio in first clinical remission rate (CR(1)) than T-LBL ones (64.4% vs 30.8%, P=0.032) . Group with WBC count higher than 50×10(9)/L at presentation showed higher ration of achieving CR(1) than those lower than 50×10(9)/L (78.4% vs 50.9%, P=0.010) . In comparison with the non-ETP-ALL, ETP-ALL patients had older age of onset (P<0.001) , lower WBC count (P<0.001) , lower risk of CNS involvement (10.0% vs 30.2%, P=0.009) and slightly inferior overall survival (P=0.073) . T-cell lineage markers CD1a(-), CD8(-) and CD4(-) positive patients had higher CR(1) than their corresponding negative ones (P=0.002, P=0.000, P=0.001) , while CD33(-) and CD56(-) positive patients had lower ratio of achieving CR(1) than their negative ones, respectively (P=0.035, P=0.035) . Conclusion: Flow cytometry and associated markers for immunophenotyping was of significance in the diagnosis and prognosis monitoring of T-ALL/LBL. The percentage of ETP-ALL/LBL subtype was high in Chinese adolescent and adult T-ALL patients. ETP-ALL/LBL was a high risk subtype, which needs more precise standard for diagnosis and advanced therapies for better outcome.


Subject(s)
Precursor Cells, T-Lymphoid/cytology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Adolescent , Adult , Aged , China , Female , Humans , Immunophenotyping , Male , Middle Aged , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/classification , Prognosis , Young Adult
11.
Chinese Journal of Hematology ; (12): 561-567, 2019.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1012187

ABSTRACT

Objective: To evaluate the clinical characteristics of T-cell acute leukemia/lymphoma (T-ALL) and explore the prognosis significance of early T-cell precursor leukemia/lymphoma. Methods: A cohort of 126 patients diagnosed with T-ALL from 2008 to 2014 in West China Hospital, Sichuan University were enrolled in this study. They were further categorized by immunophenotype according to the expression of T-cell lineage markers CD1a, CD8, CD5 and one or more stem cell or myeloid markers. The laboratory indicators and prognosis factors were also statistically analyzed. Results: Of all patients, the ratio of male to female was 2.5∶1, with the median age of 25 years old (range 14 to 77) . The percentage of ETP-ALL was up to 47.6%. T-ALL patients showed higher ratio in first clinical remission rate (CR(1)) than T-LBL ones (64.4% vs 30.8%, P=0.032) . Group with WBC count higher than 50×10(9)/L at presentation showed higher ration of achieving CR(1) than those lower than 50×10(9)/L (78.4% vs 50.9%, P=0.010) . In comparison with the non-ETP-ALL, ETP-ALL patients had older age of onset (P<0.001) , lower WBC count (P<0.001) , lower risk of CNS involvement (10.0% vs 30.2%, P=0.009) and slightly inferior overall survival (P=0.073) . T-cell lineage markers CD1a(-), CD8(-) and CD4(-) positive patients had higher CR(1) than their corresponding negative ones (P=0.002, P=0.000, P=0.001) , while CD33(-) and CD56(-) positive patients had lower ratio of achieving CR(1) than their negative ones, respectively (P=0.035, P=0.035) . Conclusion: Flow cytometry and associated markers for immunophenotyping was of significance in the diagnosis and prognosis monitoring of T-ALL/LBL. The percentage of ETP-ALL/LBL subtype was high in Chinese adolescent and adult T-ALL patients. ETP-ALL/LBL was a high risk subtype, which needs more precise standard for diagnosis and advanced therapies for better outcome.


Subject(s)
Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , China , Immunophenotyping , Precursor Cells, T-Lymphoid/cytology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Prognosis
12.
Nat Immunol ; 19(12): 1427-1440, 2018 12.
Article in English | MEDLINE | ID: mdl-30374131

ABSTRACT

Multipotent progenitor cells confirm their T cell-lineage identity in the CD4-CD8- double-negative (DN) pro-T cell DN2 stages, when expression of the essential transcription factor Bcl11b begins. In vivo and in vitro stage-specific deletions globally identified Bcl11b-controlled target genes in pro-T cells. Proteomics analysis revealed that Bcl11b associated with multiple cofactors and that its direct action was needed to recruit those cofactors to selective target sites. Regions near functionally regulated target genes showed enrichment for those sites of Bcl11b-dependent recruitment of cofactors, and deletion of individual cofactors relieved the repression of many genes normally repressed by Bcl11b. Runx1 collaborated with Bcl11b most frequently for both activation and repression. In parallel, Bcl11b indirectly regulated a subset of target genes by a gene network circuit via the transcription inhibitor Id2 (encoded by Id2) and transcription factor PLZF (encoded by Zbtb16); Id2 and Zbtb16 were directly repressed by Bcl11b, and Id2 and PLZF controlled distinct alternative programs. Thus, our study defines the molecular basis of direct and indirect Bcl11b actions that promote T cell identity and block alternative potentials.


Subject(s)
Lymphopoiesis/immunology , Precursor Cells, T-Lymphoid/immunology , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Repressor Proteins/immunology , Tumor Suppressor Proteins/immunology , Animals , Cell Differentiation/immunology , Gene Expression Regulation/immunology , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/cytology , Promyelocytic Leukemia Zinc Finger Protein/immunology
13.
Eur J Immunol ; 48(9): 1573-1579, 2018 09.
Article in English | MEDLINE | ID: mdl-29979809

ABSTRACT

Long-lived plasma cells (PCs) not only provide protective humoral immunity, they are also an essential component of the autoreactive immunologic memory that may drive chronic immune responses in systemic autoimmunity, such as systemic lupus erythematosus (SLE). The therapeutic relevance of their targeting has been demonstrated in preclinical models and severe, treatment-refractory cases of autoimmune diseases using the proteasome inhibitor bortezomib. Herein, we describe in detail the dynamic serologic changes and effects on immune effector cells in eight SLE patients receiving a median two cycles of 1.3 mg/m2 intravenous bortezomib. Upon proteasome inhibition, immunoglobulin levels gradually declined by ∼30%, associated with a significant reduction of autoantibodies, and serum complement whereas B-cell activation factor levels increased. While proteasome inhibition was associated with a significant depletion of short- and long-lived PCs in peripheral blood and bone marrow by ∼50%, including those with a distinctly mature CD19- phenotype, their precursor B cells and T cells largely remained unaffected, resulting in a rapid repopulation of short-lived PCs after bortezomib withdrawal, accompanied by increasing autoantibody levels. Collectively, these findings identify proteasome inhibitors as a promising treatment option for refractory SLE, but also indicate that PC depletion needs to be combined with targeted B-cell therapies for sustained responses in systemic autoimmunity.


Subject(s)
Autoantibodies/blood , Bortezomib/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Plasma Cells/drug effects , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, T-Lymphoid/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/therapeutic use , Complement System Proteins/metabolism , Humans , Immunoglobulins/blood , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lymphocyte Count , Plasma Cells/cytology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, T-Lymphoid/cytology
14.
J Exp Med ; 215(2): 595-610, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29343500

ABSTRACT

Multipotent hematopoietic progenitors must acquire thymus-homing capacity to initiate T lymphocyte development. Despite its importance, the transcriptional program underlying this process remains elusive. Cbfß forms transcription factor complexes with Runx proteins, and here we show that Cbfß2, encoded by an RNA splice variant of the Cbfb gene, is essential for extrathymic differentiation of T cell progenitors. Furthermore, Cbfß2 endows extrathymic progenitors with thymus-homing capacity by inducing expression of the principal thymus-homing receptor, Ccr9. This occurs via direct binding of Cbfß2 to cell type-specific enhancers, as is observed in Rorγt induction during differentiation of lymphoid tissue inducer cells by activation of an intronic enhancer. As in mice, an alternative splicing event in zebrafish generates a Cbfß2-specific mRNA, important for ccr9 expression. Thus, despite phylogenetically and ontogenetically variable sites of origin of T cell progenitors, their robust thymus-homing capacity is ensured by an evolutionarily conserved mechanism emerging from functional diversification of Runx transcription factor complexes by acquisition of a novel splice variant.


Subject(s)
Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/immunology , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology , Alternative Splicing , Animals , Cell Differentiation , Cell Lineage , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor beta Subunit/deficiency , Enhancer Elements, Genetic , Evolution, Molecular , Gene Knockdown Techniques , Mice , Mice, Knockout , Mice, Mutant Strains , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , RNA, Messenger/genetics , Receptors, CCR/genetics , Receptors, CCR/immunology , Species Specificity , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/immunology , Zebrafish , Zebrafish Proteins/deficiency
15.
Nat Biotechnol ; 35(12): 1188-1195, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29106410

ABSTRACT

Many promising targets for T-cell-based cancer immunotherapies are self-antigens. During thymic selection, T cells bearing T cell receptors (TCRs) with high affinity for self-antigen are eliminated. The affinity of the remaining low-avidity TCRs can be improved to increase their antitumor efficacy, but conventional saturation mutagenesis approaches are labor intensive, and the resulting TCRs may be cross-reactive. Here we describe the in vitro maturation and selection of mouse and human T cells on antigen-expressing feeder cells to develop higher-affinity TCRs. The approach takes advantage of natural Tcrb gene rearrangement to generate diversity in the length and composition of CDR3ß. In vitro differentiation of progenitors transduced with a known Tcra gene in the presence of antigen drives differentiation of cells with a distinct agonist-selected phenotype. We purified these cells to generate TCRß chain libraries pre-enriched for target antigen specificity. Several TCRß chains paired with a transgenic TCRα chain to produce a TCR with higher affinity than the parental TCR for target antigen, without evidence of cross-reactivity.


Subject(s)
Autoantigens/metabolism , Cell Differentiation/genetics , Precursor Cells, T-Lymphoid , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Animals , Cell Line , Flow Cytometry , Genes, T-Cell Receptor beta/genetics , Humans , Mice , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Protein Binding
16.
Eur J Immunol ; 47(1): 74-83, 2017 01.
Article in English | MEDLINE | ID: mdl-27759161

ABSTRACT

Pre-T cell receptor (TCR) signaling is required for pre-T cell survival, proliferation, and differentiation from the CD4 and CD8 double negative (DN) to the double positive (DP) stage. However, the pre-TCR signal transduction pathway is not fully understood and the signaling molecules involved have not been completely identified. Phospholipase Cγ (PLCγ) 1 is an important signaling molecule that generates two second messengers, diacylglycerol and inositol 1,4,5-trisphosphate, that are important to mediate PKC activation and intracellular Ca2+ flux in many signaling pathways. Previously, we have shown that PLCγ1 is important for TCR-mediated signaling, development and T-cell activation, but the role of PLCγ1 in pre-TCR signal transduction and pre-T cell development is not known. In this study, we demonstrated that PLCγ1 expression level in pre-T cells was comparable to that in mature T cells. Deletion of PLCγ1 prior to the pre-TCR signaling stage partially blocked the DN3 to DN4 transition and reduced thymic cellularity. We also demonstrated that deletion of PLCγ1 impaired pre-T cell proliferation without affecting cell survival. Further study showed that deficiency of PLCγ1 impaired pre-TCR mediated Ca2+ flux and Erk activation. Thus our studies demonstrate that PLCγ1 is important for pre-TCR mediated signal transduction and pre-T cell development.


Subject(s)
Cell Differentiation , Phospholipase C gamma/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Animals , Biomarkers , Calcium/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cell Survival/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Genotype , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Phospholipase C gamma/deficiency , Phospholipase C gamma/genetics , Phosphorylation , Precursor Cells, T-Lymphoid/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
17.
Immunol Lett ; 179: 9-18, 2016 11.
Article in English | MEDLINE | ID: mdl-27394700

ABSTRACT

Innate lymphoid cells (ILC) have only recently been recognized as a separate entity of the lymphoid lineage. Their subpopulations share common characteristics in terms of early development and major transcriptional circuitry with their related cousins of the T cell world. It is currently hypothesized that ILCs constitute an evolutionary older version of the lymphoid immune system. They are found at all primary entry points for pathogens such as mucosal surfaces of the lung and gastrointestinal system, the skin and the liver, which is the central contact point for pathogens that breach the intestinal barrier and enter the circulation. There, ILC contribute to the first line defense as well as to organ homeostasis. However, ILC are not only involved in classical defense tasks, but also contribute to the organogenesis of lymphoid organs as well as tissue remodeling and even stem cell regeneration. ILC may, therefore, implement different functions according to their emergence in ontogeny, their development and their final tissue location. We will review here their early development from precursors of the fetal liver and the adult bone marrow as well as their late plasticity in adaptation to their environment.


Subject(s)
Cell Plasticity , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation, Developmental , Humans , Lymphocyte Subsets/cytology , Phenotype , Precursor Cells, T-Lymphoid/cytology , Signal Transduction , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Mol Ther ; 24(7): 1216-26, 2016 08.
Article in English | MEDLINE | ID: mdl-27138041

ABSTRACT

Retroviral engineering of hematopoietic stem cell-derived precursor T-cells (preTs) opens the possibility of targeted T-cell transfer across human leukocyte antigen (HLA)-barriers. Alpharetroviral vectors exhibit a more neutral integration pattern thereby reducing the risk of insertional mutagenesis. Cord blood-derived CD34+ cells were transduced and differentiated into preTs in vitro. Two promoters, elongation-factor-1-short-form, and a myeloproliferative sarcoma virus variant in combination with two commonly used envelopes were comparatively assessed choosing enhanced green fluorescent protein or a third-generation chimeric antigen receptor (CAR) against CD123 as gene of interest. Furthermore, the inducible suicide gene iCaspase 9 has been validated. Combining the sarcoma virus-derived promoter with a modified feline endogenous retrovirus envelope glycoprotein yielded in superior transgene expression and transduction rates. Fresh and previously frozen CD34+ cells showed similar transduction and expansion rates. Transgene-positive cells did neither show proliferative impairment nor alteration in their lymphoid differentiation profile. The sarcoma virus-derived promoter only could express sufficient levels of iCaspase 9 to mediate dimerizer-induced apoptosis. Finally, the CD123 CAR was efficiently expressed in CD34+ cells and proved to be functional when expressed on differentiated T-cells. Therefore, the transduction of CD34+ cells with alpharetroviral vectors represents a feasible and potentially safer approach for stem cell-based immunotherapies for cancer.


Subject(s)
Alpharetrovirus/genetics , Fetal Blood/cytology , Genetic Engineering , Genetic Vectors/genetics , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/metabolism , Antigens, CD34/metabolism , Apoptosis , Bacterial Outer Membrane Proteins , Biomarkers , Cell Differentiation , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-3 Receptor alpha Subunit/immunology , Phenotype , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transduction, Genetic , Transgenes
19.
EBioMedicine ; 4: 50-61, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26981571

ABSTRACT

Robust, long-lasting immune responses are elicited by memory T cells that possess properties of stem cells, enabling them to persist long-term and to permanently replenish the effector pools. Thus, stem cell-like memory T (TSCM) cells are of key therapeutic value and efforts are underway to characterize TSCM cells and to identify means for their targeted induction. Here, we show that inhibition of mechanistic/mammalian Target of Rapamycin (mTOR) complex 1 (mTORC1) by rapamycin or the Wnt-ß-catenin signalling activator TWS119 in activated human naive T cells leads to the induction of TSCM cells. We show that these compounds switch T cell metabolism to fatty acid oxidation as favoured metabolic programme for TSCM cell generation. Of note, pharmacologically induced TSCM cells possess superior functional features as a long-term repopulation capacity after adoptive transfer. Furthermore, we provide insights into the transcriptome of TSCM cells. Our data identify a mechanism of pharmacological mTORC1 inhibitors, allowing us to confer stemness to human naive T cells which may be significantly relevant for the design of innovative T cell-based cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Immunologic Memory , Multiprotein Complexes/metabolism , Precursor Cells, T-Lymphoid/cytology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Humans , Lymphopoiesis , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Precursor Cells, T-Lymphoid/immunology , Pyrimidines/pharmacology , Pyrroles/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
20.
Immunol Lett ; 179: 2-8, 2016 11.
Article in English | MEDLINE | ID: mdl-26844414

ABSTRACT

The interest in innate lymphoid cells (ILC) has rapidly grown during the last decade. ILC include distinct cell types that are collectively involved in host protection against pathogens and tumor cells and in the regulation of tissue homeostasis. Studies in mice enabled a broad characterization of ILC function and of their developmental requirements. In humans all mature ILC subsets have been characterized and their role in the pathogenesis of certain disease is emerging. Nonetheless, still limited information is available on human ILC development. Indeed, only the cell precursors committed toward NK cells or ILC3 have been described. Here, we review the most recent finding on human mature ILC, discussing their tissue localization and function. Moreover, we summarize the available data regarding human ILC development.


Subject(s)
Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Subsets/cytology , Organ Specificity , Phenotype , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...