Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Eur J Immunol ; 52(1): 75-84, 2022 01.
Article in English | MEDLINE | ID: mdl-34561855

ABSTRACT

Treg therapy holds promise as a potentially curative approach to establish immune tolerance in transplantation and autoimmune disease. An outstanding question is whether therapeutic Tregs have the potential to transdifferentiate into effector T-cells and, thus, exacerbate rather than suppress immune responses. In mice, the transcription factor Helios is thought to promote Treg lineage stability in a range of inflammatory contexts. In humans, the role of Helios in Tregs is less clear, in part, due to the inability to enrich and study subsets of Helios-positive versus Helios-negative Tregs. Using an in vitro expansion system, we found that loss of high Helios expression and emergence of an intermediate Helios (Heliosmid )-expressing population correlated with Treg destabilization. We used CRISPR/Cas9 to genetically ablate Helios expression in human naive or memory Tregs and found that Helios-KO and unedited Tregs were equivalent in their suppressive function and stability in inflammation. Thus, high Helios expression is a marker, but not a driver, of human Treg stability in vitro. These data highlight the importance of monitoring Helios expression in therapeutic Treg manufacturing and provide new insight into the biological function of this transcription factor in human T-cells.


Subject(s)
Antigens, Differentiation/immunology , Gene Expression Regulation/immunology , Ikaros Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation/genetics , CRISPR-Cas Systems , Cell Line , Gene Knockout Techniques , Humans , Ikaros Transcription Factor/genetics , Mice
2.
Sci Immunol ; 6(65): eabe3454, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34826260

ABSTRACT

The Ikaros family transcription factors regulate lymphocyte development. Loss-of-function variants in IKZF1 cause primary immunodeficiency, but Ikaros family members IKZF2 and IKZF3 have not yet been associated with immunodeficiency. Here, we describe a pedigree with a heterozygous truncating variant in IKZF2, encoding the transcriptional activator and repressor Helios, which is highly expressed in regulatory T cells and effector T cells, particularly of the CD8+ T cell lineage. Protein-protein interaction analysis revealed that the variant abolished heterodimerization of Helios with Ikaros and Aiolos and also prevented Helios binding to members of the Mi-2/NuRD chromatin remodeling complex. Patients carrying the IKZF2 variant presented with a combined immunodeficiency phenotype characterized by recurrent upper respiratory infections, thrush and mucosal ulcers, and chronic lymphadenopathy. With extensive immunophenotyping, functional assays, and transcriptional analysis, we show that reduced Helios expression was associated with chronic T cell activation and increased production of proinflammatory cytokines both in effector and regulatory T cells. Lymph node histology from patients indicated dysregulated germinal center reactions. Moreover, affected individuals displayed a profound reduction in circulating MAIT cell numbers. In summary, we show that this previously undescribed loss-of-function variant in Helios leads to an immunodeficiency with signs of immune overactivation.


Subject(s)
Ikaros Transcription Factor/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , Aged , Female , Germinal Center/immunology , Humans , Ikaros Transcription Factor/blood , Ikaros Transcription Factor/genetics , Male , Middle Aged , Young Adult
3.
Sci Immunol ; 6(65): eabe3981, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34826259

ABSTRACT

Helios, a member of the Ikaros family of transcription factors, is predominantly expressed in developing thymocytes, activated T cells, and regulatory T cells (Tregs). Studies in mice have emphasized its role in maintenance of Treg immunosuppressive functions by stabilizing Foxp3 expression and silencing the Il2 locus. However, its contribution to human immune homeostasis and the precise mechanisms by which Helios regulates other T cell subsets remain unresolved. Here, we investigated a patient with recurrent respiratory infections and hypogammaglobulinemia and identified a germline homozygous missense mutation in IKZF2 encoding Helios (p.Ile325Val). We found that HeliosI325V retains DNA binding and dimerization properties but loses interaction with several partners, including epigenetic remodelers. Whereas patient Tregs showed increased IL-2 production, patient conventional T cells had decreased accessibility of the IL2 locus and consequently reduced IL-2 production. Reduced chromatin accessibility was not exclusive to the IL2 locus but involved a variety of genes associated with T cell activation. Single-cell RNA sequencing of peripheral blood mononuclear cells revealed gene expression signatures indicative of a shift toward a proinflammatory, effector-like status in patient CD8+ T cells. Moreover, patient CD4+ T cells exhibited a pronounced defect in proliferation with delayed expression of surface checkpoint inhibitors, suggesting an impaired onset of the T cell activation program. Collectively, we identified a previously uncharacterized, germline-encoded inborn error of immunity and uncovered a cell-specific defect in Helios-dependent epigenetic regulation. Binding of Helios with specific partners mediates this regulation, which is ultimately necessary for the transcriptional programs that enable T cell homeostasis in health and disease.


Subject(s)
Germ Cells/immunology , Ikaros Transcription Factor/immunology , Adolescent , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Humans , Ikaros Transcription Factor/genetics , Interleukin-2/biosynthesis , Male , Mutation, Missense , T-Lymphocytes, Regulatory/immunology
4.
Dis Markers ; 2021: 5574472, 2021.
Article in English | MEDLINE | ID: mdl-34257746

ABSTRACT

Regulatory T cells (Tregs) expressing the Foxp3 transcription factor are indispensable for the maintenance of immune system homeostasis. Tregs may lose Foxp3 expression or be reprogrammed into cells that produce proinflammatory cytokines, for example, Th1-like Tregs, Th2-like Tregs, Th17-like Tregs, and Tfh-like Tregs. Accordingly, selective therapeutic molecules that manipulate Treg lineage stability and/or functional activity might have the potential to improve aberrant immune responses in human disorders. In particular, the transcription factor Helios has emerged as an important marker and modulator of Tregs. Therefore, the current review focuses on recent findings on the expression, function, and mechanisms of Helios, as well as the patterns of Foxp3+ Tregs coexpressing Helios in various human disorders, in order to explore the potential of Helios for the improvement of many immune-related diseases. The studies were selected from PubMed using the library of the Nanjing Medical University in this review. The findings of the included studies indicate that Helios expression stabilizes the phenotype and function of Foxp3+ Tregs in certain inflammatory environments. Further, Tregs coexpressing Helios and Foxp3 were identified as a specific phenotype of stronger suppressor immune cells in both humans and animal models. Importantly, there is ample evidence that Helios-expressing Foxp3+ Tregs are relevant to various human disorders, including connective tissue diseases, infectious diseases, solid organ transplantation-related immunity, and cancer. Thus, Helios+Foxp3+CD4+ Tregs could be a valuable target in human diseases, and their potential should be explored further in the clinical setting.


Subject(s)
Autoimmune Diseases/immunology , Connective Tissue Diseases/immunology , Forkhead Transcription Factors/immunology , Ikaros Transcription Factor/immunology , Infections/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/metabolism , Biomarkers/metabolism , Connective Tissue Diseases/metabolism , Forkhead Transcription Factors/metabolism , Humans , Ikaros Transcription Factor/metabolism , Infections/metabolism , Inflammation/immunology , Inflammation/metabolism , Neoplasms/metabolism , Organ Transplantation , T-Lymphocytes, Regulatory/metabolism
5.
J Clin Immunol ; 41(6): 1241-1249, 2021 08.
Article in English | MEDLINE | ID: mdl-33855675

ABSTRACT

PURPOSE: IKAROS, encoded by IKZF1, is a member of the IKAROS family of zinc-finger transcription factors playing critical roles in lymphocyte development, differentiation, and tumor suppression. Several studies demonstrated that IKZF1 mutations affecting DNA binding or homo-/hetero-dimerization are mostly associated with common variable immunodeficiency, combined immunodeficiency, or hematologic manifestations. Herein we report a likely de novo, nonsense IKZF1 mutation (p.C182*) in a baby with low T cell receptor excision circles (TREC) identified by newborn screening testing for severe combined immunodeficiency. The patient also presented a profound B cell deficiency at birth. METHODS: Genetic, functional, immunologic, and clinical outcome data associated with this patient and her mutation were evaluated. RESULTS: Mutant p.C182* was detected in the cytoplasm of the patient's primary cells, in contrast to wild type (WT) IKAROS protein, only detected in the nucleus. Functional in vitro assessments revealed that p.C182* was less stable than WT IKAROS protein and failed to bind to its target DNA binding sequence and dimerize with WT IKAROS protein, resulting in impaired pericentromeric targeting and transcriptional repression by means of haploinsufficiency. During follow-up, while a spontaneous recovery of TREC and T cells was observed, B cells improved but not to sustained normal ranges. CONCLUSIONS: Patients with IKAROS-associated diseases can present with SCID-like TREC values through newborn screening testing. IKZF1 mutations should be added to the low TREC differential, although spontaneous recovery has to be considered.


Subject(s)
Haploinsufficiency/genetics , Ikaros Transcription Factor/genetics , Mutation/genetics , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , B-Lymphocytes/immunology , Common Variable Immunodeficiency/genetics , Common Variable Immunodeficiency/immunology , DNA/genetics , HEK293 Cells , Haploinsufficiency/immunology , Humans , Ikaros Transcription Factor/immunology , Infant, Newborn , Neonatal Screening/methods , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
6.
Mol Immunol ; 132: 236-249, 2021 04.
Article in English | MEDLINE | ID: mdl-33494935

ABSTRACT

Regulatory T lymphocytes are important targets for the treatment of acute respiratory distress syndrome (ARDS). IL-35 is a newly identified IL-12 cytokine family member that plays an important protective role in a variety of immune system diseases by regulating Treg cell differentiation; however, the role of IL-35 in the pathogenesis of ARDS is still unclear. Here, we found that IL-35 was significantly elevated in adult patients with ARDS compared to controls. Additionally, IL-35 was positively and significantly correlated with IL-6, IL-10 and the oxygenation index (PaO2/FiO2 ratio) but negatively correlated with TNF-α, IL-1ß and APACHE II score during ARDS. Moreover, the proportion of Treg/CD4+ cells in the peripheral blood of ARDS patients and the expression of NF-κB in PMBCs were significantly higher than in healthy individuals. Recombinant IL-35 improved survival in a murine model of CLP-induced ARDS. Additionally, IL-35 administration decreased the inflammatory response, as reflected by lower levels of cytokines (including IL-2, TNF-α, IL-1ß and IL-6) and less lung damage in CLP-induced ARDS. Furthermore, recombinant IL-35 reduced the apoptosis of lung tissue and the expression of NF-κB signalling in a CLP-induced ARDS model and increased the proportion of Treg cells in spleen and peripheral blood. In vitro experiments revealed that IL-35 can affect the phosphorylation of STAT5 during differentiation of naïve CD4+ T lymphocytes into Foxp3+Helios+ Tregs. Our findings suggest that IL-35 attenuates ARDS by promoting the differentiation of naïve CD4+ T cells into Foxp3+Helios+ Tregs, thereby providing a novel tool for anti-ARDS therapy.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Ikaros Transcription Factor/immunology , Interleukins/immunology , Respiratory Distress Syndrome/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Cytokines/immunology , Female , Humans , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged
7.
Stem Cells ; 39(4): 389-402, 2021 04.
Article in English | MEDLINE | ID: mdl-33400834

ABSTRACT

Hematopoietic stem cells (HSCs) are the progenitor cells that give rise to the diverse repertoire of all immune cells. As they differentiate, HSCs yield a series of cell states that undergo gradual commitment to become mature blood cells. Studies of hematopoiesis in murine models have provided critical insights about the lineage relationships among stem cells, progenitors, and mature cells, and these have guided investigations of the molecular basis for these distinct developmental stages. Primary immune deficiencies are caused by inborn errors of immunity that result in immune dysfunction and subsequent susceptibility to severe and recurrent infection(s). Over the last decade there has been a dramatic increase in the number and depth of the molecular, cellular, and clinical characterization of such genetically defined causes of immune dysfunction. Patients harboring inborn errors of immunity thus represent a unique resource to improve our understanding of the multilayered and complex mechanisms underlying lymphocyte development in humans. These breakthrough discoveries not only enable significant advances in the diagnosis of such rare and complex conditions but also provide substantial improvement in the development of personalized treatments. Here, we will discuss the clinical, cellular, and molecular phenotypes, and treatments of selected inborn errors of immunity that impede, either intrinsically or extrinsically, the development of B- or T-cells at different stages.


Subject(s)
Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Lymphopoiesis/immunology , Precision Medicine/methods , Primary Immunodeficiency Diseases/genetics , Animals , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Genetic Therapy/methods , Hematopoiesis/genetics , Hematopoiesis/immunology , Hematopoietic Stem Cells/cytology , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Lymphocytes/immunology , Lymphocytes/pathology , Lymphopoiesis/genetics , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/immunology , Primary Immunodeficiency Diseases/pathology , Primary Immunodeficiency Diseases/therapy
8.
Cell Chem Biol ; 28(6): 802-812.e6, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33333026

ABSTRACT

The recent development of successful CAR (chimeric antigen receptor) T cell therapies has been accompanied by a need to better control potentially fatal toxicities that can arise from adverse immune reactions. Here we present a ligand-controlled CAR system, based on the IKZF3 ZF2 ß-hairpin IMiD-inducible degron, which allows for the reversible control of expression levels of type I membrane proteins, including CARs. Testing this system in an established mouse xenotransplantation model for acute lymphoblastic leukemia, we validate the ability of the CAR19-degron to target and kill CD19-positive cells displaying complete control/clearance of the tumor. We also demonstrate that the activity of CAR19-degron can be regulated in vivo when dosing a US Food and Drug Administration-approved drug, lenalidomide.


Subject(s)
Ikaros Transcription Factor/immunology , Immunologic Factors/pharmacology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Adolescent , Animals , Cell Line , Cell Proliferation/drug effects , Female , Humans , Ikaros Transcription Factor/chemistry , Immunologic Factors/chemistry , Male , Mice , Mice, Congenic , Mice, Inbred NOD , Mice, SCID , Middle Aged , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Receptors, Chimeric Antigen/genetics , Young Adult
9.
Cell Rep Med ; 1(7): 100127, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33205076

ABSTRACT

Accumulation of CD103+CD8+ resident memory T (TRM) cells in human lung tumors has been associated with a favorable prognosis. However, the contribution of TRM to anti-tumor immunity and to the response to immune checkpoint blockade has not been clearly established. Using quantitative multiplex immunofluorescence on cohorts of non-small cell lung cancer patients treated with anti-PD-(L)1, we show that an increased density of CD103+CD8+ lymphocytes in immunotherapy-naive tumors is associated with greatly improved outcomes. The density of CD103+CD8+ cells increases during immunotherapy in most responder, but not in non-responder, patients. CD103+CD8+ cells co-express CD49a and CD69 and display a molecular profile characterized by the expression of PD-1 and CD39. CD103+CD8+ tumor TRM, but not CD103-CD8+ tumor-infiltrating counterparts, express Aiolos, phosphorylated STAT-3, and IL-17; demonstrate enhanced proliferation and cytotoxicity toward autologous cancer cells; and frequently display oligoclonal expansion of TCR-ß clonotypes. These results explain why CD103+CD8+ TRM are associated with better outcomes in anti-PD-(L)1-treated patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , Antigens, CD/genetics , Antigens, CD/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cytotoxicity, Immunologic/drug effects , Gene Expression Regulation , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Immunologic Memory , Immunotherapy/methods , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lymphocyte Activation/drug effects , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Phosphorylation , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Retrospective Studies , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , Survival Analysis , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
10.
Front Immunol ; 11: 1291, 2020.
Article in English | MEDLINE | ID: mdl-32655569

ABSTRACT

The zinc finger protein 36-like 2, ZFP36L2, is a member of a small family of RNA-binding proteins composed by ZFP36 (also known as tristetraprolin, TTP), ZFP36L1 and ZFP36L2 in humans, with corresponding murine orthologs. These proteins bind to adenine uridine-rich element (ARE) in the 3'untranslated region of target messenger RNA and stimulate target degradation. ZFP36 functions as an anti-inflammatory modulator in murine models of inflammatory diseases by down-regulating the production of inflammatory cytokines such as tumor necrosis factor-α. However, how ZFP36L1 and ZFP36L2 alter the function of CD4+ T cells is not completely understood. We addressed this issue by searching for the target genes of ZFP36L2 by comprehensive transcriptome analysis. We observed that ZFP36L2 is highly expressed in naïve CD4+ T cells; however, when CD4+ T cells are stimulated through their T cell receptors, ZFP36L2 expression is rapidly reduced in both humans and mice. Among CD4+ T cell populations, the expression levels of ZFP36L2 in regulatory T cells (Tregs) were significantly lower than those in naïve or effector CD4+ T cells. RNA-sequence analysis revealed that the forced expression of ZFP36L2 decreased Ikzf2 (encoding Helios) expression in Foxp3+ Tregs and inhibited the ability of induced Tregs (iTregs). ZFP36L2 directly bound to and destabilized the 3'untranslated region of Ikzf2 mRNA, which contains AU-rich elements. These results indicate that ZFP36L2 reduces the expression of Ikzf2 and suppresses iTreg function, raising the interesting possibility that the inhibition of ZFP36L2 in iTregs could be a therapeutic strategy for autoimmune diseases.


Subject(s)
DNA-Binding Proteins/biosynthesis , Gene Expression Regulation/immunology , Ikaros Transcription Factor/biosynthesis , T-Lymphocytes, Regulatory/immunology , Transcription Factors/biosynthesis , Transcription Factors/immunology , Tristetraprolin/immunology , Animals , DNA-Binding Proteins/immunology , Down-Regulation , Humans , Ikaros Transcription Factor/immunology , Mice , Transcription Factors/metabolism , Tristetraprolin/metabolism
12.
Sci Immunol ; 4(41)2019 11 22.
Article in English | MEDLINE | ID: mdl-31757834

ABSTRACT

T cell receptor (TCR) stimulation and cytokine cues drive the differentiation of CD4+ naïve T cells into effector T cell populations with distinct proinflammatory or regulatory functions. Unlike adult naïve T cells, human fetal naïve CD4+ T cells preferentially differentiate into FOXP3+ regulatory T (Treg) cells upon TCR activation independent of exogenous cytokine signaling. This cell-intrinsic predisposition for Treg differentiation is implicated in the generation of tolerance in utero; however, the underlying mechanisms remain largely unknown. Here, we identify epigenetic and transcriptional programs shared between fetal naïve T and committed Treg cells that are inactive in adult naïve T cells and show that fetal-derived induced Treg (iTreg) cells retain this transcriptional program. We show that a subset of Treg-specific enhancers is accessible in fetal naïve T cells, including two active superenhancers at Helios Helios is expressed in fetal naïve T cells but not in adult naïve T cells, and fetal iTreg cells maintain Helios expression. CRISPR-Cas9 ablation of Helios in fetal naïve T cells impaired their differentiation into iTreg cells upon TCR stimulation, reduced expression of immunosuppressive genes in fetal iTreg cells such as IL10, and increased expression of proinflammatory genes including IFNG Consequently, Helios knockout fetal iTreg cells had reduced IL-10 and increased IFN-γ cytokine production. Together, our results reveal important roles for Helios in enhancing preferential fetal Treg differentiation and fine-tuning eventual Treg function. The Treg-biased programs identified within fetal naïve T cells could potentially be used to engineer enhanced iTreg populations for adoptive cellular therapies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ikaros Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , Adult , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cells, Cultured , Humans , T-Lymphocytes, Regulatory/cytology
13.
Nat Immunol ; 20(11): 1517-1529, 2019 11.
Article in English | MEDLINE | ID: mdl-31591571

ABSTRACT

The establishment of a diverse B cell antigen receptor (BCR) repertoire by V(D)J recombination also generates autoreactive B cells. Anergy is one tolerance mechanism; it renders autoreactive B cells insensitive to stimulation by self-antigen, whereas Toll-like receptor (TLR) signaling can reactivate anergic B cells. Here, we describe a critical role of the transcription factor Ikaros in controlling BCR anergy and TLR signaling. Mice with specific deletion of Ikaros in mature B cells developed systemic autoimmunity. Ikaros regulated many anergy-associated genes, including Zfp318, which is implicated in the attenuation of BCR responsiveness by promoting immunoglobulin D expression in anergic B cells. TLR signaling was hyperactive in Ikaros-deficient B cells, which failed to upregulate feedback inhibitors of the MyD88-nuclear factor κB signaling pathway. Systemic inflammation was lost on expression of a non-self-reactive BCR or loss of MyD88 in Ikaros-deficient B cells. Thus, Ikaros acts as a guardian preventing autoimmunity by promoting BCR anergy and restraining TLR signaling.


Subject(s)
Autoimmunity/genetics , B-Lymphocytes/immunology , Clonal Anergy/genetics , Ikaros Transcription Factor/metabolism , Toll-Like Receptors/metabolism , Animals , B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Immunoglobulin D/immunology , Immunoglobulin D/metabolism , Mice , Models, Animal , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/immunology
14.
J Immunol ; 203(8): 2210-2221, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31519862

ABSTRACT

HIV-1 infection expands large populations of late-stage differentiated CD8 T cells that may persist long after viral escape from TCR recognition. In this study, we investigated whether such CD8 T cell populations can perform unconventional innate-like antiviral effector functions. Chronic untreated HIV-1 infection was associated with elevated numbers of CD45RA+CD57+ terminal effector CD8 T cells expressing FcγRIIIA (CD16). The FcγRIIIA+ CD8 T cells displayed a distinctive transcriptional profile between conventional CD8 T cells and NK cells, characterized by high levels of IKZF2 and low expression of IL7R This transcriptional profile translated into a distinct NKp80+ IL-7Rα- surface phenotype with high expression of the Helios transcription factor. Interestingly, the FcγRIIIA+ CD8 T cells mediated HIV-specific Ab-dependent cellular cytotoxicity (ADCC) activity at levels comparable with NK cells on a per cell basis. The FcγRIIIA+ CD8 T cells were highly activated in a manner that correlated positively with expansion of the CD8 T cell compartment and with plasma levels of soluble mediators of antiviral immunity and inflammation such as IP-10, TNF, IL-6, and TNFRII. The frequency of FcγRIIIA+ CD8 T cells persisted as patients initiated suppressive antiretroviral therapy, although their activation levels declined. These data indicate that terminally differentiated effector CD8 T cells acquire enhanced innate cell-like characteristics during chronic viral infection and suggest that HIV-specific ADCC is a function CD8 T cells use to target HIV-infected cells. Furthermore, as the FcγRIIIA+ CD8 T cells persist in treatment, they contribute significantly to the ADCC-capable effector cell pool in patients on antiretroviral therapy.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Ikaros Transcription Factor/immunology , Receptors, IgG/genetics , Receptors, Interleukin-7/immunology , Adolescent , Adult , Antibody-Dependent Cell Cytotoxicity/genetics , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cohort Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Receptors, IgG/immunology , Young Adult
15.
Immunology ; 158(3): 161-170, 2019 11.
Article in English | MEDLINE | ID: mdl-31517385

ABSTRACT

Regulatory T (Treg) cells are a subset of CD4+ T cells that are critical for the maintenance of self-tolerance. The forkhead box transcription factor Foxp3 is a master regulator for the Treg phenotype and function and its expression is essential in Treg cells, as the loss of Foxp3 results in lethal autoimmunity. Two major subsets of Treg cells have been described in vivo; thymus-derived Treg (tTreg) cells that develop in the thymus and peripherally induced Treg (pTreg) cells that are derived from conventional CD4+  Foxp3- T cells and are converted in peripheral tissues to cells that express Foxp3 and acquire suppressive ability. The transcription factor Helios, a member of the Ikaros transcription factor family, is expressed in 60-70% of Treg cells in both mouse and man, and is believed to be a marker of tTreg cells. In this review, we discuss the role and function of Helios in Treg cells, the controversy surrounding the use of Helios as a marker of tTreg cells, and how Helios controls specific aspects of the Treg cell program.


Subject(s)
Antigens, Differentiation/immunology , DNA-Binding Proteins/immunology , Forkhead Transcription Factors/immunology , Ikaros Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/immunology , Animals , Humans , Mice
16.
Proc Natl Acad Sci U S A ; 116(30): 15178-15183, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285322

ABSTRACT

We derived a mouse model in which a mutant form of Nbn/Nbs1mid8 (hereafter Nbnmid8) exhibits severely impaired binding to the Mre11-Rad50 core of the Mre11 complex. The Nbnmid8 allele was expressed exclusively in hematopoietic lineages (in Nbn-/mid8vav mice). Unlike Nbnflox/floxvav mice with Nbn deficiency in the bone marrow, Nbn-/mid8vav mice were viable. Nbn-/mid8vav mice hematopoiesis was profoundly defective, exhibiting reduced cellularity of thymus and bone marrow, and stage-specific blockage of B cell development. Within 6 mo, Nbn-/mid8 mice developed highly penetrant T cell leukemias. Nbn-/mid8vav leukemias recapitulated mutational features of human T cell acute lymphoblastic leukemia (T-ALL), containing mutations in NOTCH1, TP53, BCL6, BCOR, and IKZF1, suggesting that Nbnmid8 mice may provide a venue to examine the relationship between the Mre11 complex and oncogene activation in the hematopoietic compartment. Genomic analysis of Nbn-/mid8vav malignancies showed focal amplification of 9qA2, causing overexpression of MRE11 and CHK1 We propose that overexpression of MRE11 compensates for the metastable Mre11-Nbnmid8 interaction, and that selective pressure for overexpression reflects the essential role of Nbn in promoting assembly and activity of the Mre11 complex.


Subject(s)
Acid Anhydride Hydrolases/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , MRE11 Homologue Protein/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , T-Lymphocytes/immunology , Acid Anhydride Hydrolases/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/immunology , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Disease Models, Animal , Genomic Instability/immunology , Hematopoiesis/genetics , Hematopoiesis/immunology , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , MRE11 Homologue Protein/immunology , Mice , Mice, Knockout , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Protein Binding , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Repressor Proteins/genetics , Repressor Proteins/immunology , Signal Transduction , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
17.
Eur J Immunol ; 49(9): 1344-1355, 2019 09.
Article in English | MEDLINE | ID: mdl-31151137

ABSTRACT

The Ikaros family of transcription factors (TFs) are important regulators of lymphocyte function. However, their roles in human innate lymphoid cell (ILC) function remain unclear. Here, we found that Ikaros (IKZF1) is expressed by all ILC subsets, including NK cells, in blood, tonsil, and gut, while Helios (IKZF2) is preferentially expressed by ILC3 in tonsil and gut. Aiolos (IKZF3) followed the expression pattern of T-bet and Eomes, being predominantly expressed by ILC1 and NK cells. Differentiation of IFN-γ-producing ILC1 and NK cells from ILC3 by IL-1ß plus IL-12-stimulation was associated with upregulation of T-bet and Aiolos. Selective degradation of Aiolos and Ikaros by lenalidomide suppressed ILC1 and NK cell differentiation and expression of ILC1 and NK cell-related transcripts (LEF1, PRF1, GRZB, CD244, NCR3, and IRF8). In line with reduced ILC1/NK cell differentiation, we observed an increase in the expression of the ILC3-related TF Helios, as well as ILC3 transcripts (TNFSF13B, IL22, NRP1, and RORC) and in the frequency of IL-22 producing ILC3 in cultures with IL-1ß and IL-23. These data suggest that suppression of Aiolos and Ikaros expression inhibits ILC1 and NK cell differentiation while ILC3 function is maintained. Hence, our results open up for new possibilities in targeting Ikaros family TFs for modulation of type 1/3 immunity in inflammation and cancer.


Subject(s)
Cell Differentiation/immunology , Cell Transdifferentiation/immunology , Ikaros Transcription Factor/immunology , Killer Cells, Natural/immunology , Lenalidomide/immunology , Cells, Cultured , Humans , Immunity, Innate/immunology , Interferon Regulatory Factors/immunology , Interleukin-12/immunology , Interleukin-1beta/immunology , Lymphocytes/immunology
18.
Nat Commun ; 10(1): 1911, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015454

ABSTRACT

Multiple myeloma is a malignancy of antibody-secreting plasma cells. Most patients benefit from current therapies, however, 20% of patients relapse or die within two years and are deemed high risk. Here we analyze structural variants from 795 newly-diagnosed patients as part of the CoMMpass study. We report translocations involving the immunoglobulin lambda (IgL) locus are present in 10% of patients, and indicative of poor prognosis. This is particularly true for IgL-MYC translocations, which coincide with focal amplifications of enhancers at both loci. Importantly, 78% of IgL-MYC translocations co-occur with hyperdiploid disease, a marker of standard risk, suggesting that IgL-MYC-translocated myeloma is being misclassified. Patients with IgL-translocations fail to benefit from IMiDs, which target IKZF1, a transcription factor that binds the IgL enhancer at some of the highest levels in the myeloma epigenome. These data implicate IgL translocation as a driver of poor prognosis which may be due to IMiD resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Immunoglobulin lambda-Chains/genetics , Multiple Myeloma/diagnosis , Myeloma Proteins/genetics , Translocation, Genetic , Antineoplastic Agents/therapeutic use , DNA Copy Number Variations , Drug Resistance, Neoplasm/immunology , Enhancer Elements, Genetic , Genetic Loci , Genome, Human , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Immunologic Factors/therapeutic use , Lenalidomide/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Plasma Cells/immunology , Plasma Cells/pathology , Prognosis , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Recurrence , Survival Analysis , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Whole Genome Sequencing
19.
Cell Physiol Biochem ; 52(5): 1178-1192, 2019.
Article in English | MEDLINE | ID: mdl-30990587

ABSTRACT

BACKGROUND/AIMS: Rheumatoid arthritis (RA) is a progressive, chronic, even disabling systemic autoimmune disease. Imbalance between pathogenic immune cells and immunosuppressive cells is associated with the pathogenesis and development of RA and other autoimmune diseases. As Foxp3 is also expressed on activated CD4+ cells in the presence of inflammation, the identification of Treg cells in patients with RA remains a challenge. METHODS: Comprehensive analyses were carried out by Flow cytometry. Expression of Helios, CD226, T cell immunoreceptor with Ig and ITIM domains clinical samples and healthy controls. RESULTS: We have systemically examined three potential markers, Helios, CD226 and TIGIT, that are possibly related to Treg identification, and found that Helios expression on CD4+Foxp3+cells was decreased and negatively correlated with the disease activity of RA patients, while CD226 and TIGIT both showed elevated expression levels in CD4+Foxp3+cells in RA patients and they were not associated with disease activity of RA patients. CONCLUSION: Taken together, our findings indicate that CD4+CD25hiCD127low/-Foxp3+Helios+ may represent the real Treg cell population in patients with RA.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation/immunology , Arthritis, Rheumatoid/immunology , Forkhead Transcription Factors/immunology , Ikaros Transcription Factor/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Arthritis, Rheumatoid/pathology , Female , Humans , Male , Middle Aged , T-Lymphocytes, Regulatory/pathology
20.
J Immunol ; 202(4): 1112-1123, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30635395

ABSTRACT

CD4 Th cells are organizers of the immune response, directing other immune cells to initiate and maintain effective humoral and cellular immunity. CD4 T cells differentiate into distinct Th effector or regulatory subsets in response to signals delivered to them during the course of infection. Ikaros is a transcription factor that is expressed in blood cells from the level of the hematopoietic stem cell. It is required for normal thymic T cell development and serves as a tumor suppressor, as lack of Ikaros in developing lymphoid cells results in leukemia. To study the role of Ikaros in CD4 T cell differentiation and function, an Ikaros conditional knockout mouse was developed such that Ikaros expression was deleted specifically in mature T cells, thus avoiding defects observed in germline Ikaros mutant mice. Using this model system, we have shown that in the absence of Ikaros, CD4 T cells are able to attain Th1, Th2, and Th17, but not inducible regulatory T, cell fates. However, they show enhanced expression of a cohort of proinflammatory cytokines, resulting in differentiation of Th17 cells with a phenotype that has been associated with autoimmunity and pathological inflammation. In addition, we define Ikaros as a repressor of the gene program associated with the response to type I IFNs, another key pathway whose deregulation is linked to autoimmunity. Taken together, these data definitively define Ikaros as a critical regulator at the center of the inflammatory response in T cells and highlight a potential role in suppressing autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Ikaros Transcription Factor/immunology , Inflammation/immunology , Animals , Female , Ikaros Transcription Factor/deficiency , Ikaros Transcription Factor/genetics , Inflammation/genetics , Interferon Type I/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...