Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Blood ; 135(3): 167-180, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31805184

ABSTRACT

NF-κB is a key regulator of inflammation and cancer progression, with an important role in leukemogenesis. Despite its therapeutic potential, targeting NF-κB using pharmacologic inhibitors has proven challenging. Here, we describe a myeloid cell-selective NF-κB inhibitor using an miR-146a mimic oligonucleotide conjugated to a scavenger receptor/Toll-like receptor 9 agonist (C-miR146a). Unlike an unconjugated miR146a, C-miR146a was rapidly internalized and delivered to the cytoplasm of target myeloid cells and leukemic cells. C-miR146a reduced expression of classic miR-146a targets (IRAK1 and TRAF6), thereby blocking activation of NF-κB in target cells. IV injections of C-miR146a mimic to miR-146a-deficient mice prevented excessive NF-κB activation in myeloid cells, and thus alleviated myeloproliferation and mice hypersensitivity to bacterial challenge. Importantly, C-miR146a showed efficacy in dampening severe inflammation in clinically relevant models of chimeric antigen receptor (CAR) T-cell-induced cytokine release syndrome. Systemic administration of C-miR146a oligonucleotide alleviated human monocyte-dependent release of IL-1 and IL-6 in a xenotransplanted B-cell lymphoma model without affecting CD19-specific CAR T-cell antitumor activity. Beyond anti-inflammatory functions, miR-146a is a known tumor suppressor commonly deleted or expressed at reduced levels in human myeloid leukemia. Using The Cancer Genome Atlas acute myeloid leukemia data set, we found an inverse correlation of miR-146a levels with NF-κB-related genes and with patient survival. Correspondingly, C-miR146a induced cytotoxic effects in human MDSL, HL-60, and MV4-11 leukemia cells in vitro. The repeated IV administration of C-miR146a inhibited expression of NF-κB target genes and thereby thwarted progression of disseminated HL-60 leukemia. Our results show the potential of using myeloid cell-targeted miR-146a mimics for the treatment of inflammatory and myeloproliferative disorders.


Subject(s)
Cytokine Release Syndrome/prevention & control , Inflammation/prevention & control , Leukemia, Myeloid, Acute/prevention & control , MicroRNAs/genetics , Myeloid Progenitor Cells/pathology , NF-kappa B/metabolism , Animals , Apoptosis , Cell Proliferation , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/pathology , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Myeloid Progenitor Cells/metabolism , NF-kappa B/genetics , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
RNA ; 26(2): 126-136, 2020 02.
Article in English | MEDLINE | ID: mdl-31740586

ABSTRACT

At the heart of an innate immune response lies a tightly regulated gene expression program. This precise regulation is crucial because small changes can shift the balance from protective to destructive immunity. Here we identify a frequently used alternative splice site in the gene oligoadenylate synthetase 1g (Oas1g), a key component of the 2-5A antiviral system. Usage of this splice site leads to the generation of a transcript subject to decay, and removal of the site leads to increased expression of Oas1g and an improved antiviral response. However, removal of the splice site also leads to an increase in apoptotic cell death, suggesting this splicing event exists as a compromise between the pathogen protective benefits and collateral damage associated with OAS1g activity. Across the innate immune response, we show that a multitude of alternative splicing events predicted to lead to decay exist, and thus have the potential to play a significant role in the regulation of gene expression in innate immunity.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Alternative Splicing , Antiviral Agents/metabolism , Gene Expression Regulation/genetics , Immunity, Innate/genetics , RNA Splice Sites , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Apoptosis , HEK293 Cells , Humans , Mice , Nonsense Mediated mRNA Decay , RAW 264.7 Cells
3.
Cell Rep ; 25(11): 2992-3005.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540934

ABSTRACT

Long-term hematopoietic stem cells (LT-HSCs) maintain hematopoietic output throughout an animal's lifespan. However, with age, the balance is disrupted, and LT-HSCs produce a myeloid-biased output, resulting in poor immune responses to infectious challenge and the development of myeloid leukemias. Here, we show that young and aged LT-HSCs respond differently to inflammatory stress, such that aged LT-HSCs produce a cell-intrinsic, myeloid-biased expression program. Using single-cell RNA sequencing (scRNA-seq), we identify a myeloid-biased subset within the LT-HSC population (mLT-HSCs) that is prevalent among aged LT-HSCs. We identify CD61 as a marker of mLT-HSCs and show that CD61-high LT-HSCs are uniquely primed to respond to acute inflammatory challenge. We predict that several transcription factors regulate the mLT-HSCs gene program and show that Klf5, Ikzf1, and Stat3 play an important role in age-related inflammatory myeloid bias. We have therefore identified and isolated an LT-HSC subset that regulates myeloid versus lymphoid balance under inflammatory challenge and with age.


Subject(s)
Aging/pathology , Hematopoietic Stem Cells/metabolism , Inflammation/pathology , Animals , Biomarkers/metabolism , Inflammation/genetics , Ligands , Mice, Inbred C57BL , Models, Biological , Myeloid Cells/metabolism , Toll-Like Receptors/metabolism , Transcription, Genetic
4.
Nat Commun ; 9(1): 3338, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30115909

ABSTRACT

Li-Fan Lu and Alexander Y. Rudensky, who supplied miR-146a floxed mice used in this study, were inadvertently omitted from the author list in the originally published version of this Article. This has now been corrected in both the PDF and HTML versions of the Article. The generation of the floxed mice has been described in detail by Cho and Lee et al.1.

5.
Nat Commun ; 8(1): 851, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29021573

ABSTRACT

The innate inflammatory response must be tightly regulated to ensure effective immune protection. NF-κB is a key mediator of the inflammatory response, and its dysregulation has been associated with immune-related malignancies. Here, we describe a miRNA-based regulatory network that enables precise NF-κB activity in mouse macrophages. Elevated miR-155 expression potentiates NF-κB activity in miR-146a-deficient mice, leading to both an overactive acute inflammatory response and chronic inflammation. Enforced miR-155 expression overrides miR-146a-mediated repression of NF-κB activation, thus emphasizing the dominant function of miR-155 in promoting inflammation. Moreover, miR-155-deficient macrophages exhibit a suboptimal inflammatory response when exposed to low levels of inflammatory stimuli. Importantly, we demonstrate a temporal asymmetry between miR-155 and miR-146a expression during macrophage activation, which creates a combined positive and negative feedback network controlling NF-κB activity. This miRNA-based regulatory network enables a robust yet time-limited inflammatory response essential for functional immunity.MicroRNAs (miR) are important regulators of gene transcription, with miR-155 and miR-146a both implicated in macrophage activation. Here the authors show that NF-κB signalling, miR-155 and miR-146a form a complex network of cross-regulations to control gene transcription in macrophages for modulating inflammatory responses.


Subject(s)
Macrophages/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , Animals , HEK293 Cells , Humans , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism
6.
J Exp Med ; 212(10): 1679-92, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26371188

ABSTRACT

MicroRNAs have emerged as key regulators of B cell fate decisions and immune function. Deregulation of several microRNAs in B cells leads to the development of autoimmune disease and cancer in mice. We demonstrate that the microRNA-212/132 cluster (miR-212/132) is induced in B cells in response to B cell receptor signaling. Enforced expression of miR-132 results in a block in early B cell development at the prepro-B cell to pro-B cell transition and induces apoptosis in primary bone marrow B cells. Importantly, loss of miR-212/132 results in accelerated B cell recovery after antibody-mediated B cell depletion. We find that Sox4 is a target of miR-132 in B cells. Co-expression of SOX4 with miR-132 rescues the defect in B cell development from overexpression of miR-132 alone, thus suggesting that miR-132 may regulate B lymphopoiesis through Sox4. In addition, we show that the expression of miR-132 can inhibit cancer development in cells that are prone to B cell cancers, such as B cells expressing the c-Myc oncogene. We have thus uncovered miR-132 as a novel contributor to B cell development.


Subject(s)
B-Lymphocytes/physiology , MicroRNAs/genetics , SOXC Transcription Factors/genetics , 3' Untranslated Regions , Animals , Apoptosis/genetics , B-Lymphocytes/pathology , Cell Survival , Gene Expression Regulation , HEK293 Cells , Humans , Leukemia, B-Cell/genetics , Leukemia, Experimental/genetics , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Multigene Family , SOXC Transcription Factors/metabolism
7.
Immunity ; 42(6): 1021-32, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084022

ABSTRACT

MicroRNAs are critical post-transcriptional regulators of hematopoietic cell-fate decisions, though little remains known about their role in aging hematopoietic stem cells (HSCs). We found that the microRNA-212/132 cluster (Mirc19) is enriched in HSCs and is upregulated during aging. Both overexpression and deletion of microRNAs in this cluster leads to inappropriate hematopoiesis with age. Enforced expression of miR-132 in the bone marrow of mice led to rapid HSC cycling and depletion. A genetic deletion of Mirc19 in mice resulted in HSCs that had altered cycling, function, and survival in response to growth factor starvation. We found that miR-132 exerted its effect on aging HSCs by targeting the transcription factor FOXO3, a known aging associated gene. Our data demonstrate that Mirc19 plays a role in maintaining balanced hematopoietic output by buffering FOXO3 expression. We have thus identified it as a potential target that might play a role in age-related hematopoietic defects.


Subject(s)
Bone Marrow Cells/physiology , Forkhead Transcription Factors/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , MicroRNAs/metabolism , Aging/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line , Cell Survival/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Stem Cell Factor/metabolism
8.
Bone ; 79: 21-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26008608

ABSTRACT

Osteoclasts are multinucleated, bone-resorbing cells formed via fusion of monocyte progenitors, a process triggered by prolonged stimulation with RANKL, the osteoclast master regulator cytokine. Monocyte fusion into osteoclasts has been shown to play a key role in bone remodeling and homeostasis; therefore, aberrant fusion may be involved in a variety of bone diseases. Indeed, research in the last decade has led to the discovery of genes regulating osteoclast fusion; yet the basic cellular regulatory mechanism underlying the fusion process is poorly understood. Here, we applied a novel approach for tracking the fusion processes, using live-cell imaging of RANKL-stimulated and non-stimulated progenitor monocytes differentially expressing dsRED or GFP, respectively. We show that osteoclast fusion is initiated by a small (~2.4%) subset of precursors, termed "fusion founders", capable of fusing either with other founders or with non-stimulated progenitors (fusion followers), which alone, are unable to initiate fusion. Careful examination indicates that the fusion between a founder and a follower cell consists of two distinct phases: an initial pairing of the two cells, typically lasting 5-35 min, during which the cells nevertheless maintain their initial morphology; and the fusion event itself. Interestingly, during the initial pre-fusion phase, a transfer of the fluorescent reporter proteins from nucleus to nucleus was noticed, suggesting crosstalk between the founder and follower progenitors via the cytoplasm that might directly affect the fusion process, as well as overall transcriptional regulation in the developing heterokaryon.


Subject(s)
Monocytes/cytology , Osteoclasts/cytology , RANK Ligand/metabolism , Stem Cells/cytology , Animals , Bone Resorption/metabolism , Cell Differentiation/physiology , Cell Fusion , Cell Line , Coculture Techniques , Fluorescent Antibody Technique , Mice , Monocytes/metabolism , Osteoclasts/metabolism , RAW 264.7 Cells , Stem Cells/metabolism
9.
Nat Struct Mol Biol ; 19(6): 650-2, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22580560

ABSTRACT

Primary microRNA cleavage by the Drosha-Dgcr8 'Microprocessor' complex is critical for microRNA biogenesis. Yet, the Microprocessor may also cleave other nuclear RNAs in a nonspecific manner. We studied Microprocessor function using mathematical modeling and experiments in mouse and human tissues. We found that the autoregulatory feedback on Microprocessor expression is instrumental for balancing the efficiency and specificity of its activity by effectively tuning Microprocessor levels to those of its pri-miRNA substrate.


Subject(s)
MicroRNAs/metabolism , Proteins/metabolism , Animals , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Biological , Proteins/genetics , RNA-Binding Proteins
10.
Proc Natl Acad Sci U S A ; 107(36): 15804-9, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20720163

ABSTRACT

When stem cells and multipotent progenitors differentiate, they undergo fate restriction, enabling a single fate and blocking differentiation along alternative routes. We herein present a mechanism whereby such unequivocal commitment is achieved, based on microRNA (miRNA)-dependent repression of an alternative cell fate. We show that the commitment of monocyte RAW264.7 progenitors to active macrophage differentiation involves rapid up-regulation of miR-155 expression, which leads to the suppression of the alternative pathway, namely RANK ligand-induced osteoclastogenesis, by repressing the expression of MITF, a transcription factor essential for osteoclast differentiation. A temporal asymmetry, whereby miR-155 expression precedes and overrides the activation of the osteoclast transcriptional program, provides the means for coherent macrophage differentiation, even in the presence of osteoclastogenic signals. Based on these findings, we propose that miRNA may provide a general mechanism for the unequivocal commitment underlying stem cell differentiation.


Subject(s)
Cell Lineage , MicroRNAs/physiology , Stem Cells/cytology , Animals , Cell Line , Mice , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...