Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Immunity ; 54(3): 514-525.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33657395

ABSTRACT

MicroRNAs are important regulators of immune responses. Here, we show miR-221 and miR-222 modulate the intestinal Th17 cell response. Expression of miR-221 and miR-222 was induced by proinflammatory cytokines and repressed by the cytokine TGF-ß. Molecular targets of miR-221 and miR-222 included Maf and Il23r, and loss of miR-221 and miR-222 expression shifted the transcriptomic spectrum of intestinal Th17 cells to a proinflammatory signature. Although the loss of miR-221 and miR-222 was tolerated for maintaining intestinal Th17 cell homeostasis in healthy mice, Th17 cells lacking miR-221 and miR-222 expanded more efficiently in response to IL-23. Both global and T cell-specific deletion of miR-221 and miR-222 rendered mice prone to mucosal barrier damage. Collectively, these findings demonstrate that miR-221 and miR-222 are an integral part of intestinal Th17 cell response that are induced after IL-23 stimulation to constrain the magnitude of proinflammatory response.


Subject(s)
Inflammation/immunology , Interleukin-23/metabolism , Intestinal Mucosa/immunology , MicroRNAs/genetics , Th17 Cells/immunology , Animals , Feedback, Physiological , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
2.
Genes Dev ; 33(15-16): 1048-1068, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31221665

ABSTRACT

Fetal hematopoietic stem and progenitor cells (HSPCs) hold promise to cure a wide array of hematological diseases, and we previously found a role for the RNA-binding protein (RBP) Lin28b in respecifying adult HSPCs to resemble their fetal counterparts. Here we show by single-cell RNA sequencing that Lin28b alone was insufficient for complete reprogramming of gene expression from the adult toward the fetal pattern. Using proteomics and in situ analyses, we found that Lin28b (and its closely related paralog, Lin28a) directly interacted with Igf2bp3, another RBP, and their enforced co-expression in adult HSPCs reactivated fetal-like B-cell development in vivo more efficiently than either factor alone. In B-cell progenitors, Lin28b and Igf2bp3 jointly stabilized thousands of mRNAs by binding at the same sites, including those of the B-cell regulators Pax5 and Arid3a as well as Igf2bp3 mRNA itself, forming an autoregulatory loop. Our results suggest that Lin28b and Igf2bp3 are at the center of a gene regulatory network that mediates the fetal-adult hematopoietic switch. A method to efficiently generate induced fetal-like hematopoietic stem cells (ifHSCs) will facilitate basic studies of their biology and possibly pave a path toward their clinical application.


Subject(s)
Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , Hematopoietic Stem Cells/physiology , RNA-Binding Proteins/metabolism , Animals , Binding Sites , Cells, Cultured , DNA-Binding Proteins/genetics , Mice , MicroRNAs/metabolism , Models, Animal , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
3.
Nat Immunol ; 14(11): 1190-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056746

ABSTRACT

Although intergenic long noncoding RNAs (lincRNAs) have been linked to gene regulation in various tissues, little is known about lincRNA transcriptomes in the T cell lineages. Here we identified 1,524 lincRNA clusters in 42 T cell samples, from early T cell progenitors to terminally differentiated helper T cell subsets. Our analysis revealed highly dynamic and cell-specific expression patterns for lincRNAs during T cell differentiation. These lincRNAs were located in genomic regions enriched for genes that encode proteins with immunoregulatory functions. Many were bound and regulated by the key transcription factors T-bet, GATA-3, STAT4 and STAT6. We found that the lincRNA LincR-Ccr2-5'AS, together with GATA-3, was an essential component of a regulatory circuit in gene expression specific to the TH2 subset of helper T cells and was important for the migration of TH2 cells.


Subject(s)
Gene Expression Regulation/immunology , Precursor Cells, T-Lymphoid/metabolism , RNA, Long Noncoding/genetics , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Animals , Cell Differentiation , Cell Movement , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Genetic Loci , Mice , Mice, Inbred C57BL , Molecular Sequence Annotation , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Protein Binding , RNA, Long Noncoding/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Transcriptome/immunology
4.
Immunity ; 44(6): 1241-3, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332724

ABSTRACT

Like the story of Jekyll and Hyde, Th17 cells have two guises. One helps with host immunity, but the other can cause immunopathology. In this issue of Immunity, Dong and colleagues report that three microRNAs, miR-183, miR-96, and miR-182, can enhance the pathogenicity of Th17 cells (Ichiyama et al., 2016).


Subject(s)
MicroRNAs/immunology , Th17 Cells/immunology , Humans
5.
Nat Immunol ; 13(6): 587-95, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544395

ABSTRACT

Distinct CD4(+) T cell subsets are critical for host defense and immunoregulation. Although these subsets can act as terminally differentiated lineages, they have been increasingly noted to demonstrated plasticity. MicroRNAs are factors that control T cell stability and plasticity. Here we report that naturally occurring regulatory T cells (T(reg) cells) had high expression of the microRNA miR-10a and that miR-10a was induced by retinoic acid and transforming growth factor-ß (TGF-ß) in inducible T(reg) cells. By simultaneously targeting the transcriptional repressor Bcl-6 and the corepressor Ncor2, miR-10a attenuated the phenotypic conversion of inducible T(reg) cells into follicular helper T cells. We also found that miR-10a limited differentiation into the T(H)17 subset of helper T cells and therefore represents a factor that can fine-tune the plasticity and fate of helper T cells.


Subject(s)
MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Animals , Cell Differentiation/immunology , Down-Regulation/immunology , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , Nuclear Receptor Co-Repressor 2/immunology , Phenotype , Proto-Oncogene Proteins c-bcl-6/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transcription, Genetic
6.
Immunity ; 40(6): 865-79, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24856900

ABSTRACT

Specification of the T helper 17 (Th17) cell lineage requires a well-defined set of transcription factors, but how these integrate with posttranscriptional and epigenetic programs to regulate gene expression is poorly understood. Here we found defective Th17 cell cytokine expression in miR-155-deficient CD4+ T cells in vitro and in vivo. Mir155 was bound by Th17 cell transcription factors and was highly expressed during Th17 cell differentiation. miR-155-deficient Th17 and T regulatory (Treg) cells expressed increased amounts of Jarid2, a DNA-binding protein that recruits the Polycomb Repressive Complex 2 (PRC2) to chromatin. PRC2 binding to chromatin and H3K27 histone methylation was increased in miR-155-deficient cells, coinciding with failure to express Il22, Il10, Il9, and Atf3. Defects in Th17 cell cytokine expression and Treg cell homeostasis in the absence of Mir155 could be partially suppressed by Jarid2 deletion. Thus, miR-155 contributes to Th17 cell function by suppressing the inhibitory effects of Jarid2.


Subject(s)
Cytokines/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Polycomb Repressive Complex 2/immunology , Th17 Cells/immunology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Chromatin/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology
7.
Cell ; 132(5): 860-74, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18329371

ABSTRACT

To explore the role of Dicer-dependent control mechanisms in B lymphocyte development, we ablated this enzyme in early B cell progenitors. This resulted in a developmental block at the pro- to pre-B cell transition. Gene-expression profiling revealed a miR-17 approximately 92 signature in the 3'UTRs of genes upregulated in Dicer-deficient pro-B cells; a top miR-17 approximately 92 target, the proapoptotic molecule Bim, was highly upregulated. Accordingly, B cell development could be partially rescued by ablation of Bim or transgenic expression of the prosurvival protein Bcl-2. This allowed us to assess the impact of Dicer deficiency on the V(D)J recombination program in developing B cells. We found intact Ig gene rearrangements in immunoglobulin heavy (IgH) and kappa chain loci, but increased sterile transcription and usage of D(H) elements of the DSP family in IgH, and increased N sequence addition in Igkappa due to deregulated transcription of the terminal deoxynucleotidyl transferase gene.


Subject(s)
Antibody Diversity , B-Lymphocytes/cytology , Cell Survival , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , 3' Untranslated Regions/chemistry , 3' Untranslated Regions/metabolism , Animals , Blotting, Northern , Gene Expression Profiling , Gene Rearrangement, B-Lymphocyte , Immunoglobulins/genetics , Mice , Mice, Knockout , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III , Specific Pathogen-Free Organisms
8.
Proc Natl Acad Sci U S A ; 117(42): 26520-26530, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33020268

ABSTRACT

The human genome encodes for over 1,500 RNA-binding proteins (RBPs), which coordinate regulatory events on RNA transcripts. Most studies of RBPs have concentrated on their action on host protein-encoding mRNAs, which constitute a minority of the transcriptome. A widely neglected subset of our transcriptome derives from integrated retroviral elements, termed endogenous retroviruses (ERVs), that comprise ∼8% of the human genome. Some ERVs have been shown to be transcribed under physiological and pathological conditions, suggesting that sophisticated regulatory mechanisms to coordinate and prevent their ectopic expression exist. However, it is unknown how broadly RBPs and ERV transcripts directly interact to provide a posttranscriptional layer of regulation. Here, we implemented a computational pipeline to determine the correlation of expression between individual RBPs and ERVs from single-cell or bulk RNA-sequencing data. One of our top candidates for an RBP negatively regulating ERV expression was RNA-binding motif protein 4 (RBM4). We used photoactivatable ribonucleoside-enhanced cross-linking and immunoprecipitation to demonstrate that RBM4 indeed bound ERV transcripts at CGG consensus elements. Loss of RBM4 resulted in an elevated transcript level of bound ERVs of the HERV-K and -H families, as well as increased expression of HERV-K envelope protein. We pinpointed RBM4 regulation of HERV-K to a CGG-containing element that is conserved in the LTRs of HERV-K-10, -K-11, and -K-20, and validated the functionality of this site using reporter assays. In summary, we systematically identified RBPs that may regulate ERV function and demonstrate a role for RBM4 in controlling ERV expression.


Subject(s)
Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Binding Sites , Databases, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation , Genome, Human , Humans , Immunoprecipitation , RNA/genetics , RNA/metabolism , RNA-Binding Motifs , RNA-Binding Proteins/genetics , Transcriptome
9.
Immunity ; 38(4): 742-53, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601686

ABSTRACT

MicroRNAs (miRNAs) regulate the function of several immune cells, but their role in promoting CD8(+) T cell immunity remains unknown. Here we report that miRNA-155 is required for CD8(+) T cell responses to both virus and cancer. In the absence of miRNA-155, accumulation of effector CD8(+) T cells was severely reduced during acute and chronic viral infections and control of virus replication was impaired. Similarly, Mir155(-/-) CD8(+) T cells were ineffective at controlling tumor growth, whereas miRNA-155 overexpression enhanced the antitumor response. miRNA-155 deficiency resulted in accumulation of suppressor of cytokine signaling-1 (SOCS-1) causing defective cytokine signaling through STAT5. Consistently, enforced expression of SOCS-1 in CD8(+) T cells phenocopied the miRNA-155 deficiency, whereas SOCS-1 silencing augmented tumor destruction. These findings identify miRNA-155 and its target SOCS-1 as key regulators of effector CD8(+) T cells that can be modulated to potentiate immunotherapies for infectious diseases and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Melanoma, Experimental/immunology , MicroRNAs/metabolism , Adoptive Transfer , Animals , Apoptosis/genetics , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , RNA, Small Interfering/genetics , STAT6 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Virus Replication/genetics
10.
Immunol Rev ; 261(1): 23-49, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123275

ABSTRACT

The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.


Subject(s)
Gene Regulatory Networks , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation , Cell Lineage , Epigenesis, Genetic , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Humans , Immunity, Cellular , Immunity, Innate , Immunomodulation , Transcriptome
11.
J Immunol ; 194(11): 5047-52, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25980029

ABSTRACT

MicroRNAs (miRNAs) are endogenous oligoribonucleotides with exciting therapeutic potential. Early studies established a clear role for miRNAs in leukocyte biology. The first miRNA-based therapy, miravirsen, is now in phase 2 clinical trials, making the reality of these therapies undeniable. The capacity for miRNAs to fine-tune inflammatory signaling make them attractive treatment targets for immunological diseases. Nonetheless, the degree of redundancy among miRNAs, coupled with the promiscuity of miRNA binding sites in the transcriptome, require consideration when designing miRNA-directed interventions. Altered miRNA expression occurs across a range of inflammatory conditions, including inflammatory bowel disease, arthritis, and diabetes. However, very few studies successfully treated murine models of immunological diseases with miRNA-based approaches. While discussing recent studies targeting miRNAs to treat immunological conditions, we also reflect on the risks of miRNA targeting and showcase some newer delivery systems that may improve the pharmacological profile of this class of therapeutics.


Subject(s)
Immune System Diseases/drug therapy , Immune System Diseases/genetics , Inflammation/drug therapy , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy , Animals , Binding Sites/genetics , Drug Delivery Systems , Humans , Immune System Diseases/immunology , Inflammation/genetics , Inflammation/immunology , Mice , MicroRNAs/genetics , Oligonucleotides/therapeutic use
12.
Immunol Rev ; 253(1): 290-303, 2013 May.
Article in English | MEDLINE | ID: mdl-23550653

ABSTRACT

The discovery of microRNAs has renewed interest in posttranscriptional modes of regulation, fueling an emerging view of a rich RNA world within our cells that deserves further exploration. Much work has gone into elucidating genetic regulatory networks that orchestrate gene expression programs and direct cell fate decisions in the hematopoietic system. However, the focus has been to elucidate signaling pathways and transcriptional programs. To bring us one step closer to reverse engineering the molecular logic of cellular differentiation, it will be necessary to map posttranscriptional circuits as well and integrate them in the context of existing network models. In this regard, RNA-binding proteins (RBPs) may rival transcription factors as important regulators of cell fates and represent a tractable opportunity to connect the RNA world to the proteome. ChIP-seq has greatly facilitated genome-wide localization of DNA-binding proteins, helping us to understand genomic regulation at a systems level. Similarly, technological advances such as CLIP-seq allow transcriptome-wide mapping of RBP binding sites, aiding us to unravel posttranscriptional networks. Here, we review RBP-mediated posttranscriptional regulation, paying special attention to findings relevant to the immune system. As a prime example, we highlight the RBP Lin28B, which acts as a heterochronic switch between fetal and adult lymphopoiesis.


Subject(s)
DNA-Binding Proteins/immunology , Hematopoiesis , Lymphocytes/immunology , MicroRNAs/immunology , RNA-Binding Proteins/immunology , Animals , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Hematopoiesis/genetics , Humans , MicroRNAs/genetics , RNA Interference , Signal Transduction , Transcriptome
13.
J Immunol ; 193(8): 4032-42, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25217159

ABSTRACT

Differentiation of CD4(+) helper and CD8(+) cytotoxic αß T cells from CD4(+)CD8(+) thymocytes involves upregulation of lineage-specifying transcription factors and transcriptional silencing of CD8 or CD4 coreceptors, respectively, in MHC class II or I (MHCII or I)-restricted thymocytes. In this study, we demonstrate that inactivation of the Dicer RNA endonuclease in murine thymocytes impairs initiation of Cd4 and Cd8 silencing, leading to development of positively selected MHCI- and MHCII-restricted mature CD4(+)CD8(+) thymocytes. Expression of the antiapoptotic BCL2 protein or inactivation of the p53 proapoptotic protein rescues these thymocytes from apoptosis, increasing their frequency and permitting accumulation of CD4(+)CD8(+) αß T cells in the periphery. Dicer-deficient MHCI-restricted αß T cells fail to normally silence Cd4 and display impaired induction of the CD8 lineage-specifying transcription factor Runx3, whereas Dicer-deficient MHCII-restricted αß T cells show impaired Cd8 silencing and impaired induction of the CD4 lineage-specifying transcription factor Thpok. Finally, we show that the Drosha RNA endonuclease, which functions upstream of Dicer in microRNA biogenesis, also regulates Cd4 and Cd8 silencing. Our data demonstrate a previously dismissed function for the microRNA biogenesis machinery in regulating expression of lineage-specifying transcription factors and silencing of Cd4 and Cd8 during αß T cell differentiation.


Subject(s)
DEAD-box RNA Helicases/genetics , MicroRNAs/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Ribonuclease III/genetics , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Helper-Inducer/cytology , Animals , Apoptosis/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Core Binding Factor Alpha 3 Subunit/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/immunology , Tumor Suppressor Protein p53/metabolism , Up-Regulation
14.
BMC Genomics ; 16: 903, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26546038

ABSTRACT

BACKGROUND: Transcriptional enhancers are frequently bound by a set of transcription factors that collaborate to activate lineage-specific gene expression. Recently, it was appreciated that a subset of enhancers comprise extended clusters dubbed stretch- or super-enhancers (SEs). These SEs are located near key cell identity genes, and enriched for non-coding genetic variations associated with disease. Previously, SEs have been defined as having the highest density of Med1, Brd4 or H3K27ac by ChIP-seq. The histone acetyltransferase P300 has been used as a marker of enhancers, but little is known about its binding to SEs. RESULTS: We establish that P300 marks a similar SE repertoire in embryonic stem cells as previously reported using Med1 and H3K27ac. We also exemplify a role for SEs in mouse T helper cell fate decision. Similarly, upon activation of macrophages by bacterial endotoxin, we found that many SE-associated genes encode inflammatory proteins that are strongly up-regulated. These SEs arise from small, low-density enhancers in unstimulated macrophages. We also identified expression quantitative trait loci (eQTL) in human monocytes that lie within such SEs. In macrophages and Th17 cells, inflammatory SEs can be perturbed either genetically or pharmacologically thus revealing new avenues to target inflammation. CONCLUSIONS: Our findings support the notion that P300-marked SEs can help identify key nodes of transcriptional control during cell fate decisions. The SE landscape changes drastically during cell differentiation and cell activation. As these processes are crucial in immune responses, SEs may be useful in revealing novel targets for treating inflammatory diseases.


Subject(s)
E1A-Associated p300 Protein/metabolism , Animals , E1A-Associated p300 Protein/genetics , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Humans , Mice , Quantitative Trait Loci/genetics
15.
Blood ; 122(6): 1034-41, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23798711

ABSTRACT

Reactivation of fetal hemoglobin (HbF) holds therapeutic potential for sickle cell disease and ß-thalassemias. In human erythroid cells and hematopoietic organs, LIN28B and its targeted let-7 microRNA family, demonstrate regulated expression during the fetal-to-adult developmental transition. To explore the effects of LIN28B in human erythroid cell development, lentiviral transduction was used to knockdown LIN28B expression in erythroblasts cultured from human umbilical cord CD34+ cells. The subsequent reduction in LIN28B expression caused increased expression of let-7 and significantly reduced HbF expression. Conversely, LIN28B overexpression in cultured adult erythroblasts reduced the expression of let-7 and significantly increased HbF expression. Cellular maturation was maintained including enucleation. LIN28B expression in adult erythroblasts increased the expression of γ-globin, and the HbF content of the cells rose to levels >30% of their hemoglobin. Expression of carbonic anhydrase I, glucosaminyl (N-acetyl) transferase 2, and miR-96 (three additional genes marking the transition from fetal-to-adult erythropoiesis) were reduced by LIN28B expression. The transcription factor BCL11A, a well-characterized repressor of γ-globin expression, was significantly down-regulated. Independent of LIN28B, experimental suppression of let-7 also reduced BCL11A expression and significantly increased HbF expression. LIN28B expression regulates HbF levels and causes adult human erythroblasts to differentiate with a more fetal-like phenotype.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroblasts/cytology , Erythrocytes/cytology , Fetal Hemoglobin/metabolism , Gene Expression Regulation , Antigens, CD34/metabolism , Carbonic Anhydrase I/metabolism , Cell Culture Techniques , Fetal Blood/cytology , Hemoglobin A/metabolism , Humans , MicroRNAs/metabolism , N-Acetylglucosaminyltransferases/metabolism , Phenotype , RNA-Binding Proteins
16.
Adv Immunol ; 161: 85-108, 2024.
Article in English | MEDLINE | ID: mdl-38763703

ABSTRACT

Hematopoiesis, a process which generates blood and immune cells, changes significantly during mammalian development. Definitive hematopoiesis is marked by the emergence of long-term hematopoietic stem cells (HSCs). Here, we will focus on the post-transcriptional differences between fetal liver (FL) and adult bone marrow (ABM) HSCs. It remains unclear how or why exactly FL HSCs transition to ABM HSCs, but we aim to leverage their differences to revive an old idea: in utero HSC transplantation. Unexpectedly, the expression of certain RNA-binding proteins (RBPs) play an important role in HSC specification, and can be employed to convert or reprogram adult HSCs back to a fetal-like state. Among other features, FL HSCs have a broad differentiation capacity that includes the ability to regenerate both conventional B and T cells, as well as innate-like or unconventional lymphocytes such as B-1a and marginal zone B (MzB) cells. This chapter will focus on RNA binding proteins, namely LIN28B and IGF2BP3, that are expressed during fetal life and how they promote B-1a cell development. Furthermore, this chapter considers a potential clinical application of synthetic co-expression of LIN28B and IGF2BP3 in HSCs.


Subject(s)
B-Lymphocytes , Hematopoietic Stem Cells , RNA-Binding Proteins , Humans , Animals , RNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation , Hematopoiesis , RNA Processing, Post-Transcriptional , Lymphopoiesis/genetics , Hematopoietic Stem Cell Transplantation
17.
bioRxiv ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38712171

ABSTRACT

Interferon-stimulated genes (ISGs) comprise a program of immune effectors important for host immune defense. When uncontrolled, ISGs play a central role in interferonopathies and other inflammatory diseases. The mechanisms responsible for turning on ISGs are not completely known. By investigating MATRIN3 (MATR3), a nuclear RNA-binding protein mutated in familial ALS, we found that perturbing MATR3 results in elevated expression of ISGs. Using an integrative approach, we elucidate a pathway that leads to activation of cGAS-STING. This outlines a plausible mechanism for pathogenesis in a subset of ALS, and suggests new diagnostic and therapeutic approaches for this fatal disease.

18.
Noncoding RNA ; 9(6)2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38133209

ABSTRACT

Allergy is a type 2 immune reaction triggered by antigens known as allergens, including food and environmental substances such as peanuts, plant pollen, fungal spores, and the feces and debris of mites and insects. Macrophages are myeloid immune cells with phagocytic abilities that process exogenous and endogenous antigens. Upon activation, they can produce effector molecules such as cytokines as well as anti-inflammatory molecules. The dysregulation of macrophage function can lead to excessive type 1 inflammation as well as type 2 inflammation, which includes allergic reactions. Thus, it is important to better understand how macrophages are regulated in the pathogenesis of allergies. Emerging evidence highlights the role of noncoding RNAs (ncRNAs) in macrophage polarization, which in turn can modify the pathogenesis of various immune-mediated diseases, including allergies. This review summarizes the current knowledge regarding this topic and considers three classes of ncRNAs: microRNAs, long ncRNAs, and circular ncRNAs. Understanding the roles of these ncRNAs in macrophage polarization will provide new insights into the pathogenesis of allergies and identify potential novel therapeutic targets.

19.
Proc Natl Acad Sci U S A ; 106(4): 1122-7, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19164542

ABSTRACT

Dicer is central to the RNA interference (RNAi) pathway, because it is required for processing of double-stranded RNA (dsRNA) precursors into small RNA effector molecules. In principle, any long dsRNA could serve as a substrate for Dicer. The X inactive specific transcript (Xist) is an untranslated RNA that is required for dosage compensation in mammals. It coats and silences 1 of the 2 X chromosomes in female cells and initiates a chromosomewide change in chromatin structure that includes the recruitment of Polycomb proteins, but it is largely unknown how Xist RNA mediates these processes. To investigate a potential link between the RNAi pathway and X inactivation, we generated and analyzed Dicer-deficient embryonic stem (ES) cells. In the absence of Dicer, coating by Xist RNA, initiation of silencing, and recruitment of Polycomb proteins occur normally. Dicer ablation had modest effects on the steady-state levels of spliced Xist RNA. Together our data indicate that the RNAi machinery is not essential for the initiation of X inactivation.


Subject(s)
DEAD-box RNA Helicases/deficiency , Endoribonucleases/deficiency , X Chromosome Inactivation/genetics , Animals , Crosses, Genetic , DEAD-box RNA Helicases/metabolism , Doxycycline/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoribonucleases/metabolism , Female , Gene Silencing/drug effects , Genes, X-Linked , Inbreeding , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Polycomb-Group Proteins , RNA, Long Noncoding , RNA, Untranslated/metabolism , Repressor Proteins/metabolism , Ribonuclease III , Tetracycline/pharmacology , X Chromosome/genetics , X Chromosome Inactivation/drug effects
20.
Elife ; 112022 05 26.
Article in English | MEDLINE | ID: mdl-35617021

ABSTRACT

Mitotically stable random monoallelic gene expression (RME) is documented for a small percentage of autosomal genes. We developed an in vivo genetic model to study the role of enhancers in RME using high-resolution single-cell analysis of natural killer (NK) cell receptor gene expression and enhancer deletions in the mouse germline. Enhancers of the RME NK receptor genes were accessible and enriched in H3K27ac on silent and active alleles alike in cells sorted according to allelic expression status, suggesting enhancer activation and gene expression status can be decoupled. In genes with multiple enhancers, enhancer deletion reduced gene expression frequency, in one instance converting the universally expressed gene encoding NKG2D into an RME gene, recapitulating all aspects of natural RME including mitotic stability of both the active and silent states. The results support the binary model of enhancer action, and suggest that RME is a consequence of general properties of gene regulation by enhancers rather than an RME-specific epigenetic program. Therefore, many and perhaps all genes may be subject to some degree of RME. Surprisingly, this was borne out by analysis of several genes that define different major hematopoietic lineages, that were previously thought to be universally expressed within those lineages: the genes encoding NKG2D, CD45, CD8α, and Thy-1. We propose that intrinsically probabilistic gene allele regulation is a general property of enhancer-controlled gene expression, with previously documented RME representing an extreme on a broad continuum.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily K , Regulatory Sequences, Nucleic Acid , Alleles , Animals , Chromosomes , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Mice
SELECTION OF CITATIONS
SEARCH DETAIL