Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Commun ; 12(1): 2782, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986293

ABSTRACT

Chronic stimulation of CD8+ T cells triggers exhaustion, a distinct differentiation state with diminished effector function. Exhausted cells exist in multiple differentiation states, from stem-like progenitors that are the key mediators of the response to checkpoint blockade, through to terminally exhausted cells. Due to its clinical relevance, there is substantial interest in defining the pathways that control differentiation and maintenance of these subsets. Here, we show that chronic antigen induces the anergy-associated transcription factor EGR2 selectively within progenitor exhausted cells in both chronic LCMV and tumours. EGR2 enables terminal exhaustion and stabilizes the exhausted transcriptional state by both direct EGR2-dependent control of key exhaustion-associated genes, and indirect maintenance of the exhausted epigenetic state. We show that EGR2 is a regulator of exhaustion that epigenetically and transcriptionally maintains the differentiation competency of progenitor exhausted cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Clonal Anergy/immunology , Early Growth Response Protein 2/metabolism , Lymphopoiesis/physiology , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Early Growth Response Protein 2/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
BMC Cancer ; 20(1): 966, 2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33023539

ABSTRACT

BACKGROUND: Nuclear factor of activated T cells 2 (NFAT2) has been reported to regulate the development and malignancy of few tumors. In this study, we aimed to explore the effect of NFAT2 expression on cell fate of HepG2 cell and its potential mechanisms. METHODS: Firstly, the pcDNA3.1-NFAT2 plasmid was transfected into HepG2 cells to construct NFAT2 overexpressed HepG2 cells. Then, the chemical count kit-8 cell viability assay, Annexin V-FITC apoptosis detection, EdU labeling proliferation detection, transwell and wound healing experiments were performed. The expression of Egr2 and FasL, and the phosphorylation of AKT and ERK, after ionomycin and PMA co-stimulation, was detected, while the Ca2+ mobilization stimulated by K+ solution was determined. At last, the mRNA and protein expression of NFAT2, Egr2, FasL, COX-2 and c-myc in carcinoma and adjacent tissues was investigated. RESULTS: The NFAT2 overexpression suppressed the cell viability, invasion and migration capabilities, and promoted apoptosis of HepG2 cells. NFAT2 overexpression induced the expression of Egr2 and FasL and suppressed the phosphorylation of AKT and ERK. The sensitivity and Ca2+ mobilization of HepG2 cells was also inhibited by NFAT2 overexpression. Compared with adjacent tissues, the carcinoma tissues expressed less NFAT2, Egr2, FasL and more COX-2 and c-myc. CONCLUSION: The current study firstly suggested that NFAT2 suppressed the aggression and malignancy of HepG2 cells through inducing the expression of Egr2. The absence of NFAT2 and Egr2 in carcinoma tissues reminded us that NFAT2 may be a promising therapeutic target for hepatocellular carcinoma treatment.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Early Growth Response Protein 2/metabolism , Liver Neoplasms/metabolism , NFATC Transcription Factors/metabolism , Apoptosis/physiology , Calcium/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement/physiology , Cell Survival/physiology , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/genetics , Fas Ligand Protein/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Transfection , Up-Regulation
3.
Int J Exp Pathol ; 100(2): 83-93, 2019 04.
Article in English | MEDLINE | ID: mdl-31090128

ABSTRACT

Schwann cells (SCs) critically maintain the plasticity of the peripheral nervous system. Peripheral nerve injuries and infections stimulate SCs in order to retrieve homeostasis in neural tissues. Previous studies indicate that Mycobacterium leprae (ML) regulates the expression of key factors related to SC identity, suggesting that alterations in cell phenotype may be involved in the pathogenesis of neural damage in leprosy. To better understand whether ML restricts the plasticity of peripheral nerves, the present study sought to determine the expression of Krox-20, Sox-10, c-Jun and p75NTR in SC culture and mice sciatic nerves, both infected by ML Thai-53 strain. Primary SC cultures were stimulated with two different multiplicities of infection (MOI 100:1; MOI 50:1) and assessed after 7 and 14 days. Sciatic nerves of nude mice (NU-Foxn1nu ) infected with ML were evaluated after 6 and 9 months. In vitro results demonstrate downregulation of Krox-20 and Sox-10 along with the increase in p75NTR-immunolabelled cells. Concurrently, sciatic nerves of infected mice showed a significant decrease in Krox-20 and increase in p75NTR. Our results corroborate previous findings on the interference of ML in the expression of factors involved in cell maturation, favouring the maintenance of a non-myelinating phenotype in SCs, with possible implications for the repair of adult peripheral nerves.


Subject(s)
Down-Regulation , Early Growth Response Protein 2/biosynthesis , Leprosy/metabolism , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Disease Models, Animal , Leprosy/microbiology , Leprosy/pathology , Mice, Nude , Mycobacterium leprae/isolation & purification , Neuronal Plasticity/physiology , Receptors, Nerve Growth Factor/metabolism , Schwann Cells/microbiology , Schwann Cells/pathology , Sciatic Nerve/microbiology , Sciatic Nerve/pathology , Tissue Culture Techniques
4.
s.l; s.n; 2019. 10 p.
Non-conventional in English | CONASS, Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLPROD, Sec. Est. Saúde SP, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1147177

ABSTRACT

Schwann cells (SCs) critically maintain the plasticity of the peripheral nervous system. Peripheral nerve injuries and infections stimulate SCs in order to retrieve homeostasis in neural tissues. Previous studies indicate that Mycobacterium leprae (ML) regulates the expression of key factors related to SC identity, suggesting that alterations in cell phenotype may be involved in the pathogenesis of neural damage in leprosy. To better understand whether ML restricts the plasticity of peripheral nerves, the present study sought to determine the expression of Krox­20, Sox­10, c­Jun and p75NTR in SC culture and mice sciatic nerves, both infected by ML Thai­53 strain. Primary SC cultures were stimulated with two different multiplicities of infection (MOI 100:1; MOI 50:1) and assessed after 7 and 14 days. Sciatic nerves of nude mice (NU­Foxn1nu) infected with ML were evaluated after 6 and 9 months. In vitro results demonstrate downregulation of Krox­20 and Sox­10 along with the increase in p75NTR­immunolabelled cells. Concurrently, sciatic nerves of infected mice showed a significant decrease in Krox­20 and increase in p75NTR. Our results corroborate previous findings on the interference of ML in the expression of factors involved in cell maturation, favouring the maintenance of a non­myelinating phenotype in SCs, with possible implications for the repair of adult peripheral nerves(AU).


Subject(s)
Animals , Mice , Schwann Cells/microbiology , Leprosy/metabolism , Leprosy/microbiology , Mycobacterium leprae/isolation & purification , Peripheral Nerves/microbiology , Schwann Cells/metabolism , In Vitro Techniques , Down-Regulation , Receptors, Nerve Growth Factor/physiology , Early Growth Response Protein 2/biosynthesis , Neuronal Plasticity/physiology
5.
Epilepsy Behav ; 71(Pt B): 226-237, 2017 06.
Article in English | MEDLINE | ID: mdl-26775236

ABSTRACT

Genetic animal models of epilepsy are an important tool for further understanding the basic cellular mechanisms underlying epileptogenesis and for developing novel antiepileptic drugs. We conducted a comparative study of gene expression in the inferior colliculus, a nucleus that triggers audiogenic seizures, using two animal models, the Wistar audiogenic rat (WAR) and the genetic audiogenic seizure hamster (GASH:Sal). For this purpose, both models were exposed to high intensity auditory stimulation, and 60min later, the inferior colliculi were collected. As controls, intact Wistar rats and Syrian hamsters were subjected to stimulation and tissue preparation protocols identical to those performed on the experimental animals. Ribonucleic acid was isolated, and microarray analysis comparing the stimulated Wistar and WAR rats showed that the genomic profile of these animals displayed significant (fold change, |FC|≥2.0 and p<0.05) upregulation of 38 genes and downregulation of 47 genes. Comparison of gene expression profiles between stimulated control hamsters and stimulated GASH:Sal revealed the upregulation of 10 genes and the downregulation of 5 genes. Among the common genes that were altered in both models, we identified the zinc finger immediate-early growth response gene Egr3. The Egr3 protein is a transcription factor that is induced by distinct stress-elicited factors. Based on immunohistochemistry, this protein was expressed in the cochlear nucleus complex, the inferior colliculus, and the hippocampus of both animal models as well as in lymphoma tumors of the GASH:Sal. Our results support that the overexpression of the Egr3 gene in both models might contribute to neuronal viability and development of lymphoma in response to stress associated with audiogenic seizures. This article is part of a Special Issue entitled "Genetic and Reflex Epilepsies, Audiogenic Seizures and Strains: From Experimental Models to the Clinic".


Subject(s)
Acoustic Stimulation/adverse effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/genetics , Epilepsy, Reflex/genetics , Seizures/genetics , Animals , Cricetinae , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 3/biosynthesis , Epilepsy, Reflex/drug therapy , Epilepsy, Reflex/metabolism , Gene Expression , Genes, Immediate-Early/genetics , Genetic Predisposition to Disease/genetics , Hippocampus/metabolism , Male , Mesocricetus , Rats , Rats, Wistar , Rodentia , Seizures/drug therapy , Seizures/metabolism , Species Specificity
6.
Biochimie ; 133: 1-6, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27889568

ABSTRACT

Expression of many miRNAs is altered in different cancers and these changes are thought to play a key role in formation and progression of cancer. In chronic myelogenous leukemia (CML) a number of miRNAs are known to be down regulated as compared to normal cells. In this report we have investigated the mechanism of this down regulation by using PMA induced differentiation of CML cell line K562 to megakaryocytes as an experimental system. On treatment with PMA, expression of many down regulated miRNAs including miR-22 is induced. PMA also induces expression of several transcription factors, including FosB, EGR1 and EGR2. Our results using a number of approaches, such as promoter reporter assay, FosB knock down and Chip assay, suggest that the expression of miR-22 is regulated transcriptionally by FosB.


Subject(s)
Cell Differentiation/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MicroRNAs/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Cell Differentiation/drug effects , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Megakaryocytes/drug effects , MicroRNAs/genetics , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Polymethacrylic Acids/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis
7.
BMC Anesthesiol ; 16(1): 95, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27737635

ABSTRACT

BACKGROUND: Propofol is a widely used intravenous anesthetic that is well-known for its protective effect in various human and animal disease models. However, the effects of propofol on neurogenesis, especially on the development of neural stem cells (NSCs), remains unknown. Related microRNAs may act as important regulators in this process. METHODS: Published Gene Expression Omnibus (GEO) DataSets related to propofol were selected and re-analyzed to screen neural development-related genes and predict microRNA (miRNA) expression using bioinformatic methods. Screening of the genes and miRNAs was then validated by qRT-PCR analysis of propofol-treated primary embryonic NSCs. RESULTS: Four differentially expressed mRNAs were identified in the screen and 19 miRNAs were predicted based on a published GEO DataSet. Two of four mRNAs and four of 19 predicted miRNAs were validated by qRT-PCR analysis of propofol-treated NSCs. Rno-miR-19a (Rno, Rattus Norvegicus) and rno-miR-137, and their target gene EGR2, as well as rno-miR-19b-2 and rno-miR-214 and their target gene ARC were found to be closely related to neural developmental processes, including proliferation, differentiation, and maturation of NSCs. CONCLUSION: Propofol influences miRNA expression; however, further studies are required to elucidate the mechanism underlying the effects of propofol on the four miRNAs and their target genes identified in this study. In particular, the influence of propofol on the entire development process of NSCs remains to be clarified.


Subject(s)
Gene Expression Regulation/drug effects , MicroRNAs/biosynthesis , MicroRNAs/drug effects , Neural Stem Cells/metabolism , Propofol/pharmacology , Animals , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/drug effects , Databases, Genetic , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/drug effects , Neural Stem Cells/drug effects , Neurogenesis/genetics , Primary Cell Culture , Rats
8.
Mol Med Rep ; 14(5): 4309-4314, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27667480

ABSTRACT

Down syndrome (DS) is the most common birth defect in children. To investigate the mechanisms of DS, the present study analyzed the bisulfite­sequencing (seq) data GSE42144, which was downloaded from the Gene Expression Omnibus. GSE42144 included DNA methylation data of three DS samples and three control samples, and RNA­seq data of two DS samples and five control samples. The methylated sites in the bisulfite­seq data were detected using Bismark and Bowtie2. The BiSeq tool was applied to determine differentially methylated regions and to identify adjacent genes. Using the Database for Annotation, Visualization and Integrated Discovery, the functions of the abnormal demethylated genes were predicted by functional enrichment analyses. Differentially expressed genes (DEGs) were then screened using a paired t­test. Furthermore, the interactions of the proteins encoded by selected genes were determined using the Search Tool for the Retrieval of Interacting Genes, and a protein­protein interaction (PPI) network was constructed using Cytoscape. A total of 74 CpG regions showed significant differential DNA methylation between the DS and normal samples. There were five abnormal demethylated DNA regions in chromosome 21. In the DS samples, a total of 43 adjacent genes were identified with demethylation in their promoter regions and one adjacent gene was identified with upregulated methylation in its promoter regions. In addition, 584 upregulated genes were identified, including 24 genes with transcriptional regulatory function. In particular, upregulated Runt­related transcription factor 1 (RUNX1) was located on chromosome 21. Functional enrichment analysis indicated that inhibitor of DNA binding 4 (ID4) was involved in neuronal differentiation and transcriptional suppression. In the PPI network, genes may be involved in DS by interacting with others, including nuclear receptor subfamily 4 group A member 2 (NR4A2)­early growth response (EGR)2 and NR4A2­EGR3. Therefore, RUNX1, NR4A2, EGR2, EGR3 and ID4 may be key genes associated with the pathogenesis of DS.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Down Syndrome/genetics , Early Growth Response Protein 2/genetics , Early Growth Response Protein 3/genetics , Inhibitor of Differentiation Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Computational Biology , Core Binding Factor Alpha 2 Subunit/biosynthesis , DNA Methylation/genetics , Databases, Genetic , Down Syndrome/pathology , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 3/biosynthesis , Gene Expression Regulation , Gene Regulatory Networks , Humans , Inhibitor of Differentiation Proteins/biosynthesis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Protein Interaction Maps/genetics , Sequence Analysis, RNA , Signal Transduction
9.
Hum Mol Genet ; 25(14): 3055-3069, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27288457

ABSTRACT

Schwann cells are myelinating glia in the peripheral nervous system that form the myelin sheath. A major cause of peripheral neuropathy is a copy number variant involving the Peripheral Myelin Protein 22 (PMP22) gene, which is located within a 1.4-Mb duplication on chromosome 17 associated with the most common form of Charcot-Marie-Tooth Disease (CMT1A). Rodent models of CMT1A have been used to show that reducing Pmp22 overexpression mitigates several aspects of a CMT1A-related phenotype. Mechanistic studies of Pmp22 regulation identified enhancers regulated by the Sox10 (SRY sex determining region Y-box 10) and Egr2/Krox20 (Early growth response protein 2) transcription factors in myelinated nerves. However, relatively little is known regarding how other transcription factors induce Pmp22 expression during Schwann cell development and myelination. Here, we examined Pmp22 enhancers as a function of cell type-specificity, nerve injury and development. While Pmp22 enhancers marked by active histone modifications were lost or remodeled after injury, we found that these enhancers were permissive in early development prior to Pmp22 upregulation. Pmp22 enhancers contain binding motifs for TEA domain (Tead) transcription factors of the Hippo signaling pathway. We discovered that Tead1 and co-activators Yap and Taz are required for Pmp22 expression, as well as for the expression of Egr2 Tead1 directly binds Pmp22 and Egr2 enhancers early in development and Tead1 binding is induced during myelination, correlating with Pmp22 expression. The data identify Tead1 as a novel regulator of Pmp22 expression during development in concert with Sox10 and Egr2.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , DNA-Binding Proteins/genetics , Early Growth Response Protein 2/genetics , Myelin Proteins/genetics , Peripheral Nervous System Diseases/genetics , SOXE Transcription Factors/genetics , Transcription Factors/genetics , Animals , Charcot-Marie-Tooth Disease/pathology , DNA Copy Number Variations/genetics , DNA-Binding Proteins/biosynthesis , Disease Models, Animal , Early Growth Response Protein 2/biosynthesis , Gene Expression Regulation/genetics , Humans , Mice , Myelin Sheath/genetics , Myelin Sheath/pathology , Neurogenesis/genetics , Peripheral Nervous System Diseases/pathology , Phenotype , Schwann Cells/metabolism , Schwann Cells/pathology , TEA Domain Transcription Factors , Transcription Factors/biosynthesis
10.
J Immunol ; 196(3): 1102-7, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26718337

ABSTRACT

Guillain-Barré syndrome (GBS) is an immune-mediated peripheral neuropathy. The goal of this research was the identification of biomarkers associated with recovery from GBS. In this study, we compared the transcriptome of PBMCs from a GBS patient and her healthy twin to discover possible correlates of disease progression and recovery. The study was then extended using GBS and spinal cord injury unrelated patients with similar medications and healthy individuals. The early growth response gene-2 (EGR2) was upregulated in GBS patients during disease recovery. The results provided evidence for the implication of EGR2 in GBS and suggested a role for EGR2 in the regulation of IL-17, IL-22, IL-28A, and TNF-ß cytokines in GBS patients. These results identified biomarkers associated with GBS recovery and suggested that EGR2 overexpression has a pivotal role in the downregulation of cytokines implicated in the pathophysiology of this acute neuropathy.


Subject(s)
Biomarkers/analysis , Early Growth Response Protein 2/biosynthesis , Guillain-Barre Syndrome/immunology , Adult , Aged , Aged, 80 and over , Cytokines/biosynthesis , Early Growth Response Protein 2/immunology , Female , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Recovery of Function , Transcriptome , Up-Regulation , Young Adult
11.
J Immunol ; 195(11): 5272-84, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26519537

ABSTRACT

NKT cells constitute a small population of T cells developed in the thymus that produce large amounts of cytokines and chemokines in response to lipid Ags. Signaling through the Vα14-Jα18 TCR instructs commitment to the NKT cell lineage, but the precise signaling mechanisms that instruct their lineage choice are unclear. In this article, we report that the cytoskeletal remodeling protein, p21-activated kinase 2 (Pak2), was essential for NKT cell development. Loss of Pak2 in T cells reduced stage III NKT cells in the thymus and periphery. Among different NKT cell subsets, Pak2 was necessary for the generation and function of NKT1 and NKT2 cells, but not NKT17 cells. Mechanistically, expression of Egr2 and promyelocytic leukemia zinc finger (PLZF), two key transcription factors for acquiring the NKT cell fate, were markedly diminished in the absence of Pak2. Diminished expression of Egr2 and PLZF were not caused by aberrant TCR signaling, as determined using a Nur77-GFP reporter, but were likely due to impaired induction and maintenance of signaling lymphocyte activation molecule 6 expression, a TCR costimulatory receptor required for NKT cell development. These data suggest that Pak2 controls thymic NKT cell development by providing a signal that links Egr2 to induce PLZF, in part by regulating signaling lymphocyte activation molecule 6 expression.


Subject(s)
Antigens, CD/biosynthesis , Early Growth Response Protein 2/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , Natural Killer T-Cells/immunology , Receptors, Cell Surface/biosynthesis , p21-Activated Kinases/metabolism , Animals , Cell Differentiation/immunology , Green Fluorescent Proteins/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Thymus Gland/immunology , p21-Activated Kinases/genetics
12.
Neurochem Res ; 39(5): 875-82, 2014 May.
Article in English | MEDLINE | ID: mdl-24647971

ABSTRACT

We previously reported that social isolation (SI) rearing of rodents not only elicits a variety of behavioral abnormalities including attention deficit hyperactivity disorder-like behaviors, but also impairs fear memory in mice. This study aimed to clarify a putative mechanism underlying SI-induced conditioned fear memory deficit. Mice were group-housed (GH) or socially isolated for 2 weeks or more before the experiments. SI animals acquired contextual and auditory fear memory elucidated at 90 min and 4 h after training, respectively; however, they showed significantly impaired contextual and auditory memory performance at 24 h and 4 days after the training, respectively, indicating SI-induced deficit of the consolidation process of fear memory. Neurochemical studies conducted after behavioral tests revealed that SI mice had a significantly down-regulated level of Egr-1 but not Egr-2 in the hippocampal and cortical cytosolic fractions compared with those levels in the GH control animals. Moreover, in the SI group, phosphorylated levels of synaptic plasticity-related signaling proteins in the hippocampus, NR1 subunit of N-methyl-D-aspartate receptor, glutamate receptor 1, and calmodulin-dependent kinase II but not cyclic AMP-responsive element binding protein were significantly down-regulated compared with those levels in GH animals, whereas non-phosphorylated levels of these proteins were not affected by SI. These findings suggest that dysfunctions of Egr-1 and neuro-signaling systems are involved in SI-induced deficits of fear memory consolidation in mice.


Subject(s)
Early Growth Response Protein 1/biosynthesis , Memory Disorders/physiopathology , Social Isolation , Animals , Behavior, Animal , Cerebral Cortex/metabolism , Conditioning, Classical , Down-Regulation , Early Growth Response Protein 2/biosynthesis , Fear , Hippocampus/metabolism , Male , Memory/physiology , Mice, Inbred ICR , Neuronal Plasticity , Signal Transduction , Stress, Psychological
13.
J Immunol ; 192(4): 1972-81, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24465013

ABSTRACT

Strategies for cancer immunotherapy include activating immune system for therapeutic benefit or blockade of immune checkpoints. To harness innate immunity to fight cancer, α-galactosylceramide (α-GalCer) has been used to activate NKT cells. Unfortunately, administration of α-GalCer causes long-term NKT cell anergy, but the molecular mechanism is unclear. In this study, we showed that α-GalCer-triggered egr2/3, which induced programmed death 1 and cbl-b in NKT cells, leading to NKT cell anergy. We also uncovered the induction of the immunosuppressive myeloid-derived suppressor cells (MDSCs) in the spleen by α-GalCer that might attenuate its antitumor efficacy. The accumulation of MDSC was accompanied by 20-fold rise in their arg-1 mRNAs and enhanced expression of programmed death 1/programmed death ligand 1. Furthermore, α-GalCer-induced egr-2/3 in hepatic NKT cells upregulated their TRAIL in addition to Fas ligand (FasL) and induced alarm signaling molecule IL-33 in Kupffer cells, presumably because of liver damage triggered by TRAIL/FasL. We further demonstrated that IL-33-stimulated macrophages produce G-CSF, which in turn, boosted MDSCs. Thus, α-GalCer-induced FasL/TRAIL and IL-33 provided a novel mechanism underlying α-GalCer-induced hepatotoxicity and MDSC accumulation. In contrast, analogs of α-GalCer containing phenyl group in the lipid tail could neither induce NKT anergy nor enhance MDSCs accumulation. Furthermore, tumor-infiltrating MDSCs in mice injected repeatedly with α-GalCer were 2-fold higher than those treated with phenyl-glycolipids. These results not only revealed the induction of MDSC via IL-33 as a new mechanism for α-GalCer-elicited immunosuppression but also provided one of the mechanisms underlying the superior antitumor potency of phenyl-glycolipids. Our findings have important implications for the development of NKT-stimulatory glycolipids as vaccine adjuvants and anticancer therapeutics.


Subject(s)
B7-H1 Antigen/metabolism , Galactosylceramides/immunology , Myeloid Cells/immunology , Natural Killer T-Cells/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Arginase/genetics , B7-H1 Antigen/biosynthesis , Cell Line, Tumor , Clonal Anergy/immunology , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 3/biosynthesis , Fas Ligand Protein/biosynthesis , Female , Galactosylceramides/therapeutic use , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/metabolism , Immunosuppression Therapy , Immunotherapy , Interleukin-33 , Interleukins/metabolism , Kupffer Cells/metabolism , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Natural Killer T-Cells/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Proto-Oncogene Proteins c-cbl/biosynthesis , RNA, Messenger/biosynthesis , Spleen/immunology , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Up-Regulation
14.
Int J Dev Biol ; 57(9-10): 779-86, 2013.
Article in English | MEDLINE | ID: mdl-24307296

ABSTRACT

Neural induction and anteroposterior neural patterning occur simultaneously during Xenopus gastrulation by the inhibition of BMP and Wnt signaling, respectively. However, other processes might be necessary for determining the neural-epidermal boundary. Xenopus nodal-related-3 (Xnr3) is expressed in dorsal blastula and plays a role in neural formation. In this study, we analyzed how Xnr3 affects neural patterning to identify novel mechanisms of neural-epidermal-boundary determination. In situ hybridization revealed that ventro-animal injection with Xnr3 shifted the lateral krox20 expression domain anteriorly and reduced Otx2 expression. The mature region of Xnr3 is necessary for these effects to occur, and the pro-region accelerated them. Phalloidin labeling revealed that cells around the neural-epidermal boundary lost their slender shape following Xnr3 injection. Moreover, we analyzed the cell migration of ectodermal cells and found specific Xnr3-induced effects at the neural-epidermal boundary. These findings together suggested that Xnr3 affects anterior ectoderm migration around the neural-epidermal boundary to induce a specific neural pattern abnormality. Change of the shape of surrounding ectodermal cells and the specific migratory pattern might therefore reflect the novel mechanism of neural-epidermal boundary.


Subject(s)
Body Patterning/genetics , Brain/embryology , Epidermis/embryology , Neurulation/genetics , Transforming Growth Factor beta/genetics , Xenopus Proteins/genetics , Animals , Cell Movement/genetics , Early Growth Response Protein 2/biosynthesis , Ectoderm/embryology , Embryo, Nonmammalian/metabolism , Embryonic Induction/genetics , Eye Proteins , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Homeodomain Proteins/biosynthesis , Morpholinos , Neural Plate/cytology , Otx Transcription Factors/biosynthesis , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/metabolism , Xenopus Proteins/biosynthesis , Xenopus Proteins/metabolism , Xenopus laevis
15.
Mol Cell ; 52(3): 314-24, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24207025

ABSTRACT

Lysine acetylation regulates transcription by targeting histones and nonhistone proteins. Here we report that the central regulator of transcription, RNA polymerase II, is subject to acetylation in mammalian cells. Acetylation occurs at eight lysines within the C-terminal domain (CTD) of the largest polymerase subunit and is mediated by p300/KAT3B. CTD acetylation is specifically enriched downstream of the transcription start sites of polymerase-occupied genes genome-wide, indicating a role in early stages of transcription initiation or elongation. Mutation of lysines or p300 inhibitor treatment causes the loss of epidermal growth-factor-induced expression of c-Fos and Egr2, immediate-early genes with promoter-proximally paused polymerases, but does not affect expression or polymerase occupancy at housekeeping genes. Our studies identify acetylation as a new modification of the mammalian RNA polymerase II required for the induction of growth factor response genes.


Subject(s)
Histones/genetics , Lysine/genetics , RNA Polymerase II/metabolism , Transcription, Genetic , Acetylation , Animals , Early Growth Response Protein 2/biosynthesis , Embryonic Stem Cells/cytology , Gene Expression Regulation , Genes, fos/genetics , Histones/metabolism , Humans , Promoter Regions, Genetic , RNA Polymerase II/genetics , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
16.
Int J Mol Sci ; 14(8): 16226-39, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23924943

ABSTRACT

Gastric cancer (GC) is one of the most common cancers, with high incidences in East Asia. microRNAs (miRNAs) play essential roles in the carcinogenesis of GC. miR-20a was elevated in GC, while the potential function of miR-20a was poorly understood. miR-20a expression was examined in GC tissues and cell lines. The effects of miR-20a on the growth, migration, invasion, and chemoresistance of GC cells were examined. Luciferase reporter assay and Western blot were used to screen the target of miR-20a. miR-20a was increased in GC tissues and cell lines. miR-20a promoted the growth, migration and invasion of GC cells, enhanced the chemoresistance of GC cells to cisplatin and docetaxel. Luciferase activity and Western blot confirmed that miR-20a negatively regulated EGR2 expression. Overexpression of EGR2 significantly attenuated the oncogenic effect of miR-20a. miR-20a was involved in the carcinogenesis of GC through modulation of the EGR2 signaling pathway.


Subject(s)
Carcinogenesis/genetics , Early Growth Response Protein 2/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/genetics , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cisplatin/pharmacology , Docetaxel , Drug Resistance, Neoplasm/genetics , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/genetics , Female , Gene Expression , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , RNA, Messenger/biosynthesis , Stomach Neoplasms/metabolism , Taxoids/pharmacology
17.
J Neurosci ; 33(15): 6679-90, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23575864

ABSTRACT

Several transcription factors are essential for terminal differentiation of myelinating glia, among them the high-mobility-group-domain-containing protein Sox10. To better understand how these factors exert their effects and shape glial expression programs, we identified and characterized a physical and functional link between Sox10 and the Med12 subunit of the Mediator complex that serves as a conserved multiprotein interphase between transcription factors and the general transcription machinery. We found that Sox10 bound with two of its conserved domains to the C-terminal region of Med12 and its close relative, Med12-like. In contrast to Med12-like, substantial amounts of Med12 were detected in both Schwann cells and oligodendrocytes. Its conditional glia-specific deletion in mice led to terminal differentiation defects that were highly reminiscent of those obtained after Sox10 deletion. In support of a functional cooperation, both proteins were jointly required for Krox20 induction and were physically associated with the critical regulatory region of the Krox20 gene in myelinating Schwann cells. We conclude that Sox10 functions during terminal differentiation of myelinating glia, at least in part by Med12-dependent recruitment of the Mediator complex.


Subject(s)
Cell Differentiation/physiology , Mediator Complex/physiology , Oligodendroglia/cytology , SOXE Transcription Factors/physiology , Schwann Cells/cytology , Animals , Cell Differentiation/genetics , Cell Line , Early Growth Response Protein 2/biosynthesis , Female , Gene Expression Regulation, Developmental/genetics , Humans , Male , Mediator Complex/genetics , Mice , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Protein Binding/genetics , Protein Binding/physiology , SOXE Transcription Factors/genetics , Schwann Cells/metabolism
18.
J Biol Chem ; 288(21): 14906-16, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23536186

ABSTRACT

Deregulated gene expression in B cells often results in various lymphoid malignancies and immune deficiencies. Therefore, understanding signal-induced gene regulatory pathways involved during B cell activation is important to tackle pathologies associated with altered B cell function. Primary response genes (PRGs) are rapidly induced upon signaling in B cells and other cell types and often encode oncogenic transcription factors, which are associated with various malignancies. However, an important issue that remains unclear is whether the fundamental mechanism of activation of these genes is essentially the same under such diverse conditions. c-fos is a PRG that is induced rapidly upon activation of B cells in response to a wide variety of stimuli. Using the c-fos gene as a candidate PRG, we addressed here how it is regulated in response to tumor-promoting and antigen-mimicking signals. Our results show that although the mRNA was induced and extinguished within minutes in response to both signals, surprisingly, apparently full-length unspliced pre-mRNA persisted for several hours in both cases. However, although the mitogenic signal resulted in a more sustained mRNA response that persisted for 4 h, antigenic signaling resulted in a more robust but very transient response that lasted for <1 h. Moreover, the pre-mRNA profile exhibited significant differences between the two signals. Additionally, the splicing regulation was also observed with egr-2, but not with c-myc. Together, these results suggest a previously underappreciated regulatory step in PRG expression in B cells.


Subject(s)
B-Lymphocytes/metabolism , Early Growth Response Protein 2/biosynthesis , Gene Expression Regulation/physiology , Mitosis/physiology , Proto-Oncogene Proteins c-fyn/biosynthesis , Signal Transduction/physiology , Animals , B-Lymphocytes/cytology , Cell Line, Tumor , Early Growth Response Protein 2/genetics , Male , Mice , Proto-Oncogene Proteins c-fyn/genetics , RNA Precursors/biosynthesis , RNA Precursors/genetics , RNA Splicing/physiology , Time Factors
19.
J Immunol ; 190(1): 58-65, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23203924

ABSTRACT

Early growth response gene (Egr)-2 is important for the maintenance of T cell homeostasis and controls the development of autoimmune disease. However, the underlying mechanisms are unknown. We have now discovered that Egr-2, which is induced by TGF-ß and IL-6, negatively regulates the expression of IL-17, but not IL-2 or IFN-γ, in effector T cells. In the absence of Egr-2, CD4 T cells produce high levels of Th17 cytokines, which renders mice susceptible to experimental autoimmune encephalomyelitis induction. T cells lacking Egr-2 show increased propensity for Th17, but not Th1 or Th2, differentiation. Control of IL-17 expression and Th17 differentiation by Egr-2 is due to inhibition of Batf, a transcription factor that regulates IL-17 expression and Th17 differentiation. Egr-2 interacts with Batf in CD4 T cells and suppresses its interaction with DNA sequences derived from the IL-17 promoter, whereas the activation of STAT3 and expression of retinoic acid-related orphan receptor γt are unchanged in Th17 cells in the absence of Egr-2. Thus, Egr-2 plays an important role to intrinsically control Th17 differentiation. We also found that CD4 T cells from multiple sclerosis patients have reduced expression of Egr-2 and increased expression of IL-17 following stimulation with anti-CD3 in vitro. Collectively, our results demonstrate that Egr-2 is an intrinsic regulator that controls Th17 differentiation by inhibiting Batf activation, which may be important for the control of multiple sclerosis development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/biosynthesis , Cell Differentiation/immunology , Down-Regulation/immunology , Early Growth Response Protein 2/physiology , Feedback, Physiological/physiology , Interleukin-17/biosynthesis , Th17 Cells/immunology , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/physiology , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/deficiency , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/cytology , Th17 Cells/metabolism
20.
J Neurosci ; 32(21): 7158-68, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623660

ABSTRACT

Physical damage to the peripheral nerves triggers Schwann cell injury response in the distal nerves in an event termed Wallerian degeneration: the Schwann cells degrade their myelin sheaths and dedifferentiate, reverting to a phenotype that supports axon regeneration and nerve repair. The molecular mechanisms regulating Schwann cell plasticity in the PNS remain to be elucidated. Using both in vivo and in vitro models for peripheral nerve injury, here we show that inhibition of p38 mitogen-activated protein kinase (MAPK) activity in mice blocks Schwann cell demyelination and dedifferentiation following nerve injury, suggesting that the kinase mediates the injury signal that triggers distal Schwann cell injury response. In myelinating cocultures, p38 MAPK also mediates myelin breakdown induced by Schwann cell growth factors, such as neuregulin and FGF-2. Furthermore, ectopic activation of p38 MAPK is sufficient to induce myelin breakdown and drives differentiated Schwann cells to acquire phenotypic features of immature Schwann cells. We also show that p38 MAPK concomitantly functions as a negative regulator of Schwann cell differentiation: enforced p38 MAPK activation blocks cAMP-induced expression of Krox 20 and myelin proteins, but induces expression of c-Jun. As expected of its role as a negative signal for myelination, inhibition of p38 MAPK in cocultures promotes myelin formation by increasing the number as well as the length of individual myelin segments. Altogether, our data identify p38 MAPK as an important regulator of Schwann cell plasticity and differentiation.


Subject(s)
Cell Differentiation/physiology , Nerve Fibers, Myelinated/physiology , Schwann Cells/metabolism , Schwann Cells/physiology , Wallerian Degeneration/physiopathology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Coculture Techniques , Early Growth Response Protein 2/biosynthesis , Female , Fibroblast Growth Factor 2/pharmacology , JNK Mitogen-Activated Protein Kinases/biosynthesis , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/metabolism , Neuregulin-1/pharmacology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Rats , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Wallerian Degeneration/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...