Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Nature ; 628(8007): 408-415, 2024 Apr.
Article En | MEDLINE | ID: mdl-38480883

During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20-60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches-precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)-on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.


Efferocytosis , Macrophages , RNA Polymerase II , Transcription Elongation, Genetic , Animals , Humans , Male , Mice , Apoptosis , Cytoskeleton/metabolism , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Efferocytosis/genetics , Hydrogen-Ion Concentration , Macrophages/immunology , Macrophages/metabolism , Neurons/metabolism , Phagosomes/metabolism , RNA Polymerase II/metabolism , Transcription Factors/genetics , Zebrafish/embryology , Zebrafish/genetics , Time Factors
2.
Oncogene ; 39(36): 5839-5854, 2020 09.
Article En | MEDLINE | ID: mdl-32796959

Identification of pro-metastatic genomic alterations is urgently needed to help understand and prevent the fatal course of prostate cancer. Here, we found that the transcription factor EGR3, located at chromosome 8p21.3, is a critical metastasis suppressor. Aberrant deletion of EGR3 was found in up to 59.76% (deep deletions, 16.87%; shallow deletions, 42.89%) of prostate cancer patients. In informatics analysis, EGR3 loss was associated with prostate cancer progression and low survival rates. EGR3 expression inversely correlated with the expressions of epithelial-to-mesenchymal transition (EMT) and metastasis-related gene sets in prostate cancer tissues. In prostate cancer cells, EGR3 blocked the EMT process and suppressed cell migration and invasion. In a mouse model for cancer metastasis, EGR3 overexpression significantly suppressed bone metastases of PC3 and 22Rv1 prostate cancer cells. Mechanistically, EGR3 transcriptionally activated ZFP36, GADD45B, and SOCS3 genes by directly binding to their promoter regions. The EMT-inhibitory and tumor-suppressive roles of the EGR3 downstream genes were identified through in vitro and in silico analyses. Together, our results showed that EGR3 may be a biomarker to predict clinical outcomes and that it plays an important role in the metastatic progression of prostate cancer.


Early Growth Response Protein 3/deficiency , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement , DNA Copy Number Variations , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Follow-Up Studies , Gene Deletion , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Male , Mice , Neoplasm Metastasis , Prognosis , Proportional Hazards Models , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Risk Factors , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 113(50): E8131-E8140, 2016 12 13.
Article En | MEDLINE | ID: mdl-27911796

Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease characterized by multiorgan inflammation induced by autoantibodies. Early growth response gene 2 (Egr2), a transcription factor essential for T-cell anergy induction, controls systemic autoimmunity in mice and humans. We have previously identified a subpopulation of CD4+ regulatory T cells, CD4+CD25-LAG3+ cells, that characteristically express both Egr2 and LAG3 and control mice model of lupus via TGF-ß3 production. However, due to the mild phenotype of lymphocyte-specific Egr2-deficient mice, the presence of an additional regulator has been speculated. Here, we show that Egr2 and Egr3 expressed in T cells cooperatively prevent humoral immune responses by supporting TGF-ß3 secretion. T cell-specific Egr2/Egr3 double-deficient (Egr2/3DKO) mice spontaneously developed an early onset lupus-like disease that was more severe than in T cell-specific Egr2-deficient mice. In accordance with the observation that CD4+CD25-LAG3+ cells from Egr2/3DKO mice completely lost the capacity to produce TGF-ß3, the excessive germinal center reaction in Egr2/3DKO mice was suppressed by the adoptive transfer of WT CD4+CD25-LAG3+ cells or treatment with a TGF-ß3-expressing vector. Intriguingly, latent TGF-ß binding protein (Ltbp)3 expression maintained by Egr2 and Egr3 was required for TGF-ß3 production from CD4+CD25-LAG3+ cells. Because Egr2 and Egr3 did not demonstrate cell intrinsic suppression of the development of follicular helper T cells, Egr2- and Egr3-dependent TGF-ß3 production by CD4+CD25-LAG3+ cells is critical for controlling excessive B-cell responses. The unique attributes of Egr2/Egr3 in T cells may provide an opportunity for developing novel therapeutics for autoantibody-mediated diseases including SLE.


Autoimmunity , Early Growth Response Protein 2/immunology , Early Growth Response Protein 3/immunology , Latent TGF-beta Binding Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta3/biosynthesis , Adaptive Immunity , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Differentiation , Disease Models, Animal , Early Growth Response Protein 2/deficiency , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/pathology , Humans , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/pathology
4.
J Comp Neurol ; 524(9): 1892-919, 2016 Jun 15.
Article En | MEDLINE | ID: mdl-26660356

Motor function in mammalian species depends on the maturation of spinal circuits formed by a large variety of interneurons that regulate motoneuron firing and motor output. Interneuron activity is in turn modulated by the organization of their synaptic inputs, but the principles governing the development of specific synaptic architectures unique to each premotor interneuron are unknown. For example, Renshaw cells receive, at least in the neonate, convergent inputs from sensory afferents (likely Ia) and motor axons, raising the question of whether they interact during Renshaw cell development. In other well-studied neurons, such as Purkinje cells, heterosynaptic competition between inputs from different sources shapes synaptic organization. To examine the possibility that sensory afferents modulate synaptic maturation on developing Renshaw cells, we used three animal models in which afferent inputs in the ventral horn are dramatically reduced (ER81(-/-) knockout), weakened (Egr3(-/-) knockout), or strengthened (mlcNT3(+/-) transgenic). We demonstrate that increasing the strength of sensory inputs on Renshaw cells prevents their deselection and reduces motor axon synaptic density, and, in contrast, absent or diminished sensory afferent inputs correlate with increased densities of motor axons synapses. No effects were observed on other glutamatergic inputs. We conclude that the early strength of Ia synapses influences their maintenance or weakening during later development and that heterosynaptic influences from sensory synapses during early development regulates the density and organization of motor inputs on mature Renshaw cells.


Afferent Pathways/physiology , Gene Expression Regulation, Developmental/physiology , Motor Neurons/cytology , Renshaw Cells/physiology , Synapses/physiology , Animals , Animals, Newborn , Axons/metabolism , Calbindins/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Parvalbumins/metabolism , Phosphopyruvate Hydratase/metabolism , Spinal Cord/cytology , Synapses/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
5.
Immunity ; 37(4): 685-96, 2012 Oct 19.
Article En | MEDLINE | ID: mdl-23021953

Lymphocytes provide optimal responses against pathogens with minimal inflammatory pathology. However, the intrinsic mechanisms regulating these responses are unknown. Here, we report that deletion of both transcription factors Egr2 and Egr3 in lymphocytes resulted in a lethal autoimmune syndrome with excessive serum proinflammatory cytokines but also impaired antigen receptor-induced proliferation of B and T cells. Egr2- and Egr3-defective B and T cells had hyperactive signal transducer and activator of transcription-1 (STAT1) and STAT3 while antigen receptor-induced activation of transcription factor AP-1 was severely impaired. We discovered that Egr2 and/or Egr3 directly induced expression of suppressor of cytokine signaling-1 (SOCS1) and SOCS3, inhibitors of STAT1 and STAT3, and also blocked the function of Batf, an AP-1 inhibitor, in B and T cells. Thus, Egr2 and Egr3 regulate B and T cell function in adaptive immune responses and homeostasis by promoting antigen receptor signaling and controlling inflammation.


B-Lymphocytes/immunology , Cell Proliferation , Early Growth Response Protein 2/immunology , Early Growth Response Protein 3/immunology , T-Lymphocytes/immunology , Animals , Antigens/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , Early Growth Response Protein 2/deficiency , Early Growth Response Protein 3/deficiency , Homeostasis , Inflammation/immunology , Mice , Mice, Knockout , Signal Transduction , T-Lymphocytes/cytology , Transcription Factor AP-1/immunology
6.
Neuropsychopharmacology ; 37(10): 2285-98, 2012 Sep.
Article En | MEDLINE | ID: mdl-22692564

The immediate-early gene early growth response 3 (Egr3) is associated with schizophrenia and expressed at reduced levels in postmortem patients' brains. We have previously reported that Egr3-deficient (Egr3(-/-)) mice display reduced sensitivity to the sedating effects of clozapine compared with wild-type (WT) littermates, paralleling the heightened tolerance of schizophrenia patients to antipsychotic side effects. In this study, we have used a pharmacological dissection approach to identify a neurotransmitter receptor defect in Egr3(-/-) mice that may mediate their resistance to the locomotor suppressive effects of clozapine. We report that this response is specific to second-generation antipsychotic agents (SGAs), as first-generation medications suppress the locomotor activity of Egr3(-/-) and WT mice to a similar degree. Further, in contrast to the leading theory that sedation by clozapine results from anti-histaminergic effects, we show that H1 histamine receptors are not responsible for this effect in C57BL/6 mice. Instead, selective serotonin 2A receptor (5HT(2A)R) antagonists ketanserin and MDL-11939 replicate the effect of SGAs, repressing the activity in WT mice at a dosage that fails to suppress the activity of Egr3(-/-) mice. Radioligand binding revealed nearly 70% reduction in 5HT(2A)R expression in the prefrontal cortex of Egr3(-/-) mice compared with controls. Egr3(-/-) mice also exhibit a decreased head-twitch response to 5HT(2A)R agonist 1-(2,5-dimethoxy 4-iodophenyl)-2-amino propane (DOI). These findings provide a mechanism to explain the reduced sensitivity of Egr3(-/-) mice to the locomotor suppressive effects of SGAs, and suggest that 5HT(2A)Rs may also contribute to the sedating properties of these medications in humans. Moreover, as the deficit in cortical 5HT(2A)R in Egr3(-/-) mice aligns with numerous studies reporting decreased 5HT(2A)R levels in the brains of schizophrenia patients, and the gene encoding the 5HT(2A)R is itself a leading schizophrenia candidate gene, these findings suggest a potential mechanism by which putative dysfunction in EGR3 in humans may influence risk for schizophrenia.


Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Hypnotics and Sedatives/metabolism , Motor Activity/drug effects , Receptor, Serotonin, 5-HT2A/metabolism , Animals , Clozapine/pharmacology , Disease Models, Animal , Humans , Ketanserin , Male , Mice , Mice, Transgenic , Piperidines , Schizophrenia/metabolism , Serotonin 5-HT2 Receptor Agonists , Serotonin Antagonists/pharmacology
7.
PLoS One ; 6(9): e25696, 2011.
Article En | MEDLINE | ID: mdl-21980528

Nerve Growth Factor (NGF) is a target tissue derived neurotrophin required for normal sympathetic neuron survival and target tissue innervation. NGF signaling regulates gene expression in sympathetic neurons, which in turn mediates critical aspects of neuron survival, axon extension and terminal axon branching during sympathetic nervous system (SNS) development. Egr3 is a transcription factor regulated by NGF signaling in sympathetic neurons that is essential for normal SNS development. Germline Egr3-deficient mice have physiologic dysautonomia characterized by apoptotic sympathetic neuron death and abnormal innervation to many target tissues. The extent to which sympathetic innervation abnormalities in the absence of Egr3 is caused by altered innervation or by neuron death during development is unknown. Using Bax-deficient mice to abrogate apoptotic sympathetic neuron death in vivo, we show that Egr3 has an essential role in target tissue innervation in the absence of neuron death. Sympathetic target tissue innervation is abnormal in many target tissues in the absence of neuron death, and like NGF, Egr3 also appears to effect target tissue innervation heterogeneously. In some tissues, such as heart, spleen, bowel, kidney, pineal gland and the eye, Egr3 is essential for normal innervation, whereas in other tissues such as lung, stomach, pancreas and liver, Egr3 appears to have little role in innervation. Moreover, in salivary glands and heart, two tissues where Egr3 has an essential role in sympathetic innervation, NGF and NT-3 are expressed normally in the absence of Egr3 indicating that abnormal target tissue innervation is not due to deregulation of these neurotrophins in target tissues. Taken together, these results clearly demonstrate a role for Egr3 in mediating sympathetic target tissue innervation that is independent of neuron survival or neurotrophin deregulation.


Early Growth Response Protein 3/metabolism , Neurons/cytology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Animals , Atrophy/genetics , Atrophy/metabolism , Cell Death/genetics , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Gene Deletion , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Nerve Growth Factor/metabolism , Nerve Growth Factors/metabolism , Neurons/pathology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics
8.
Development ; 135(17): 2949-57, 2008 Sep.
Article En | MEDLINE | ID: mdl-18653557

Sympathetic nervous system development depends upon many factors that mediate neuron migration, differentiation and survival. Target tissue-derived nerve growth factor (NGF) signaling-induced gene expression is required for survival, differentiation and target tissue innervation of post-migratory sympathetic neurons. However, the transcriptional regulatory mechanisms mediated by NGF signaling are very poorly defined. Here, we identify Egr3, a member of the early growth response (Egr) family of transcriptional regulators, as having an important role in sympathetic nervous system development. Egr3 is regulated by NGF signaling and it is expressed in sympathetic neurons during development when they depend upon NGF for survival and target tissue innervation. Egr3-deficient mice have severe sympathetic target tissue innervation abnormalities and profound physiological dysautonomia. Unlike NGF, which is essential for sympathetic neuron survival and for axon branching within target tissues, Egr3 is required for normal terminal axon extension and branching, but not for neuron survival. The results indicate that Egr3 is a novel NGF signaling effector that regulates sympathetic neuron gene expression required for normal target tissue innervation and function. Egr3-deficient mice have a phenotype that is remarkably similar to humans with sympathetic nervous system disease, raising the possibility that it may have a role in some forms of human dysautonomia, most of which have no known cause.


Autonomic Nervous System Diseases/congenital , Early Growth Response Protein 3/deficiency , Sympathetic Nervous System/embryology , Sympathetic Nervous System/physiopathology , Animals , Animals, Newborn , Apoptosis/drug effects , Arylalkylamine N-Acetyltransferase/metabolism , Axons/drug effects , Axons/metabolism , Blepharoptosis/pathology , Blepharoptosis/physiopathology , Cell Movement/drug effects , Early Growth Response Protein 3/genetics , Early Growth Response Protein 3/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Gene Expression Regulation, Developmental/drug effects , Heart/innervation , Mice , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/pathology , Pineal Gland/drug effects , Pineal Gland/enzymology , Pineal Gland/innervation , Signal Transduction/drug effects , Sympathetic Nervous System/cytology , Sympathetic Nervous System/drug effects
9.
Eur J Immunol ; 38(2): 528-36, 2008 Feb.
Article En | MEDLINE | ID: mdl-18203138

TCR-induced NF-AT activation leads to the up-regulation of multiple genes involved in T cell anergy. Since NF-AT is also involved in T cell activation, we have endeavored to dissect TCR-induced activating and inhibitory genetic programs. This approach revealed roles for the early growth response (Egr) family of transcription factors and the Egr coactivator/corepressor NGFI-A-binding protein (NAB)2 in regulating T cell function. TCR-induced Egr-1 and NAB2 enhance T cell function, while Egr-2 and Egr-3 inhibit T cell function. In this report, we demonstrate that Egr-2 and Egr-3 are induced by NF-AT in the absence of AP-1, while Egr-1 and NAB2 both require AP-1-mediated transcription. Our data suggest that Egr-3 is upstream of Egr-2, and that mechanistically Egr-2 and Egr-3 suppress Egr-1 and NAB2 expression. Functionally, T cells from Egr-2 and Egr-3 null mice are hyperresponsive while T cells from Egr-3 transgenic, overexpressing mice are hyporesponsive. Furthermore, an in vivo model of autoimmune pneumonitis reveals that T cells from Egr-3 null mice hasten death while Egr-3-overexpressing T cells cause less disease. Overall, our data suggest that just as the Egr/NAB network of genes control cell fate in other systems, TCR-induced Egr-1, 2, 3 and NAB2 control the fate of antigen recognition in T cells.


Early Growth Response Protein 1/physiology , Early Growth Response Protein 2/physiology , Early Growth Response Protein 3/physiology , Neoplasm Proteins/physiology , Repressor Proteins/physiology , T-Lymphocytes/immunology , Animals , Early Growth Response Protein 1/antagonists & inhibitors , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/deficiency , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/biosynthesis , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, Antigen, T-Cell/physiology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , T-Lymphocytes/metabolism
10.
Mol Cell Biol ; 25(23): 10286-300, 2005 Dec.
Article En | MEDLINE | ID: mdl-16287845

Early growth response (Egr) transcription factors (Egr1 to Egr4) are synaptic activity-inducible immediate early genes (IEGs) that regulate some aspects of synaptic plasticity-related to learning and memory, yet the target genes regulated by them are unknown. In particular, Egr1 is essential for persistence of late-phase long-term potentiation (L-LTP), for hippocampus-dependent long-term memory formation, and for reconsolidation of previously established memories. Here, we show that Egr1 and Egr3 directly regulate the plasticity-associated activity-regulated cytoskeletal-related (Arc) gene, a synaptic activity-induced effector molecule which is also required for L-LTP and hippocampus-dependent learning and memory processing. Moreover, Egr1-deficient and Egr3-deficient mice lack Arc protein in a subpopulation of neurons, while mice lacking both Egr1 and Egr3 lack Arc in all neurons. Thus, Egr1 and Egr3 can indirectly modulate synaptic plasticity by directly regulating Arc and the plasticity mechanisms it mediates in recently activated synapses.


Cytoskeletal Proteins/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 3/metabolism , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Transcription, Genetic/genetics , Animals , Base Sequence , Cytoskeletal Proteins/metabolism , Early Growth Response Protein 1/deficiency , Early Growth Response Protein 1/genetics , Early Growth Response Protein 3/deficiency , Early Growth Response Protein 3/genetics , Gene Expression Regulation , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Kainic Acid/pharmacology , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Response Elements/genetics , Seizures/genetics , Seizures/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Synapses/drug effects , Synapses/metabolism
...