Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33622787

ABSTRACT

HLA-C arose during evolution of pregnancy in the great apes 10 to 15 million years ago. It has a dual function on placental extravillous trophoblasts (EVTs) as it contributes to both tolerance and immunity at the maternal-fetal interface. The mode of its regulation is of considerable interest in connection with the biology of pregnancy and pregnancy abnormalities. First-trimester primary EVTs in which HLA-C is highly expressed, as well as JEG3, an EVT model cell line, were employed. Single-cell RNA-seq data and quantitative PCR identified high expression of the transcription factor ELF3 in those cells. Chromatin immunoprecipitation (ChIP)-PCR confirmed that both ELF3 and MED1 bound to the proximal HLA-C promoter region. However, binding of RFX5 to this region was absent or severely reduced, and the adjacent HLA-B locus remained closed. Expression of HLA-C was inhibited by ELF3 small interfering RNAs (siRNAs) and by wrenchnolol treatment. Wrenchnolol is a cell-permeable synthetic organic molecule that mimics ELF3 and is relatively specific for binding to ELF3's coactivator, MED23, as our data also showed in JEG3. Moreover, the ELF3 gene is regulated by a superenhancer that spans more than 5 Mb, identified by assay for transposase-accessible chromatin using sequencing (ATAC-seq), as well as by its sensitivity to (+)-JQ1 (inhibitor of BRD4). ELF3 bound to its own promoter, thus creating an autoregulatory feedback loop that establishes expression of ELF3 and HLA-C in trophoblasts. Wrenchnolol blocked binding of MED23 to ELF3, thus disrupting the positive-feedback loop that drives ELF3 expression, with down-regulation of HLA-C expression as a consequence.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Feedback, Physiological , HLA-C Antigens/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Trophoblasts/immunology , Abortion, Legal , Adamantane/pharmacology , Azepines/pharmacology , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Female , Gene Expression Regulation, Developmental/immunology , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-C Antigens/immunology , Humans , Immunity, Maternally-Acquired , Indoles/pharmacology , Mediator Complex/genetics , Mediator Complex/immunology , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/immunology , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/immunology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Triazoles/pharmacology , Trophoblasts/cytology , Trophoblasts/drug effects
2.
Inflamm Res ; 70(1): 89-98, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33185705

ABSTRACT

OBJECTIVE: The dysfunction of pulmonary microvascular endothelial cells (PMVECs) is one of the critical characteristics of acute lung injury/acute respiratory distress syndrome (ALI/ARDS) induced by severe infection. PIM1 is a constitutively active serine/threonine kinase that is involved in multiple biological processes. However, the underlying correlation between PIM1 and PMVECs injury remains unclear. The main purpose of this study was to reveal roles of PIM1 and explore the potential mechanisms during the development of endotoxin-induced ALI induced by intraperitoneal LPS administration. MATERIALS AND METHODS: PIM1 level in the lung tissues of endotoxin-induced ALI mice or plasma derived from cardiopulmonary bypass (CPB)-induced ALI patients were measured. The protective roles of PIM1 specific inhibitor SMI-4a on endotoxin-induced lung injuries were evaluated through histological, permeability, neutrophil infiltration and survival assessment. The relationship between PIM1 and ELK3/ICAM-1 axis was validated in vivo and vitro. The correlation between plasma PIM1 and indicative vascular endothelium injury biomarkers (PaO2/FiO2 ratio, Ang-II, E-selectin and PAI-1) levels derived from CPB-induced ALI patient were analyzed. RESULTS: PIM1 expression in the lung tissues was increased in the mice of endotoxin-induced ALI. The PIM1 specific inhibitor SMI-4a administration relieved the severity of endotoxin-induced ALI. More importantly, PIM1 modulates ICAM1 expression through regulating transcription factor ELK3 expression in vitro. Eventually, plasma PIM1 level was positively correlated with Ang-II and PAI-1 levels but negatively correlated with SpO2/FiO2 ratio among CPB induced ALI patients. CONCLUSION: Our results indicated that PIM1 inhibition carried a protective role against endotoxin-induced ALI by modulating the ELK3/ICAM1 axis on PMVECs. PIM1 may be a potential therapeutic target for endotoxin-induced ALI.


Subject(s)
Acute Lung Injury/immunology , Endothelial Cells/immunology , Intercellular Adhesion Molecule-1/immunology , Lung/immunology , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins c-pim-1/immunology , Acute Lung Injury/chemically induced , Animals , Cardiopulmonary Bypass/adverse effects , Cells, Cultured , Humans , Lipopolysaccharides , Lung/cytology , Male , Mice, Inbred C57BL , Microvessels/cytology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/blood
3.
Nat Commun ; 11(1): 4225, 2020 08 24.
Article in English | MEDLINE | ID: mdl-32839463

ABSTRACT

Gallbladder cancer (GBC) is an aggressive gastrointestinal malignancy with no approved targeted therapy. Here, we analyze exomes (n = 160), transcriptomes (n = 115), and low pass whole genomes (n = 146) from 167 gallbladder cancers (GBCs) from patients in Korea, India and Chile. In addition, we also sequence samples from 39 GBC high-risk patients and detect evidence of early cancer-related genomic lesions. Among the several significantly mutated genes not previously linked to GBC are ETS domain genes ELF3 and EHF, CTNNB1, APC, NSD1, KAT8, STK11 and NFE2L2. A majority of ELF3 alterations are frame-shift mutations that result in several cancer-specific neoantigens that activate T-cells indicating that they are cancer vaccine candidates. In addition, we identify recurrent alterations in KEAP1/NFE2L2 and WNT pathway in GBC. Taken together, these define multiple targetable therapeutic interventions opportunities for GBC treatment and management.


Subject(s)
DNA-Binding Proteins/genetics , Frameshift Mutation , Gallbladder Neoplasms/genetics , Genetic Predisposition to Disease/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Chile , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Gallbladder Neoplasms/diagnosis , Gallbladder Neoplasms/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genomics/methods , Humans , India , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins c-ets/metabolism , Republic of Korea , Transcription Factors/immunology , Transcription Factors/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
4.
Int J Mol Sci ; 20(15)2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31370265

ABSTRACT

Osteosarcoma and Ewing sarcoma are the most common malignant primary bone tumors mainly occurring in children, adolescents and young adults. Current standard therapy includes multidrug chemotherapy and/or radiation specifically for Ewing sarcoma, associated with tumor resection. However, patient survival has not evolved for the past decade and remains closely related to the response of tumor cells to chemotherapy, reaching around 75% at 5 years for patients with localized forms of osteosarcoma or Ewing sarcoma but less than 30% in metastatic diseases and patients resistant to initial chemotherapy. Despite Ewing sarcoma being characterized by specific EWSR1-ETS gene fusions resulting in oncogenic transcription factors, currently, no targeted therapy could be implemented. It seems even more difficult to develop a targeted therapeutic strategy in osteosarcoma which is characterized by high complexity and heterogeneity in genomic alterations. Nevertheless, the common point between these different bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Therefore, targeting different actors of the bone tumor microenvironment has been hypothesized to develop new therapeutic strategies. In this context, it is well known that the Wnt/ß-catenin signaling pathway plays a key role in cancer development, including osteosarcoma and Ewing sarcoma as well as in bone remodeling. Moreover, recent studies highlight the implication of the Wnt/ß-catenin pathway in angiogenesis and immuno-surveillance, two key mechanisms involved in metastatic dissemination. This review focuses on the role played by this signaling pathway in the development of primary bone tumors and the modulation of their specific microenvironment.


Subject(s)
Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Osteosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Tumor Microenvironment/drug effects , Adolescent , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Bone and Bones , Child , Humans , Lymphatic Metastasis , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/prevention & control , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/immunology , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/mortality , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , RNA-Binding Protein EWS/antagonists & inhibitors , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/immunology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/immunology , Sarcoma, Ewing/mortality , Survival Analysis , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Wnt Signaling Pathway/drug effects , Young Adult , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/immunology
5.
J Exp Med ; 215(9): 2265-2278, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30087163

ABSTRACT

An IRF8-dependent subset of conventional dendritic cells (cDCs), termed cDC1, effectively cross-primes CD8+ T cells and facilitates tumor-specific T cell responses. Etv6 is an ETS family transcription factor that controls hematopoietic stem and progenitor cell (HSPC) function and thrombopoiesis. We report that like HSPCs, cDCs express Etv6, but not its antagonist, ETS1, whereas interferon-producing plasmacytoid dendritic cells (pDCs) express both factors. Deletion of Etv6 in the bone marrow impaired the generation of cDC1-like cells in vitro and abolished the expression of signature marker CD8α on cDC1 in vivo. Moreover, Etv6-deficient primary cDC1 showed a partial reduction of cDC-specific and cDC1-specific gene expression and chromatin signatures and an aberrant up-regulation of pDC-specific signatures. Accordingly, DC-specific Etv6 deletion impaired CD8+ T cell cross-priming and the generation of tumor antigen-specific CD8+ T cells. Thus, Etv6 optimizes the resolution of cDC1 and pDC expression programs and the functional fitness of cDC1, thereby facilitating T cell cross-priming and tumor-specific responses.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunity, Cellular , Neoplasms/immunology , Proto-Oncogene Proteins c-ets/immunology , Repressor Proteins/immunology , Animals , Antigens, Neoplasm/genetics , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/pathology , Gene Deletion , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/immunology , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombopoiesis/genetics , Thrombopoiesis/immunology , ETS Translocation Variant 6 Protein
6.
Cell Physiol Biochem ; 45(6): 2401-2410, 2018.
Article in English | MEDLINE | ID: mdl-29550824

ABSTRACT

BACKGROUND/AIMS: The E74-like factor 3 (ELF3) is an inflammatory mediator that participates in cartilage destruction in osteoarthritis. Leptin and other adipokines negatively impact articular cartilage, triggering catabolic and inflammatory responses in chondrocytes. Here, we investigated whether leptin induces ELF3 expression in chondrocytes and the signaling pathway involved in this process. METHODS: We determined mRNA and protein levels of ELF3 by RT-qPCR and Western blotting using cultured human primary chondrocytes and the human T/C-28a2 chondrocyte cell line. Further, we measured luciferase activities of different reporter constructs, and we assessed the contribution of leptin to the induction of ELF3 mRNA by knocking down hLEPR gene expression using siRNA technology. RESULTS: Leptin synergizes with IL-1ß in inducing ELF3 expression in chondrocytes. We also found that PI3K, p38, and JAK2 signaling pathways are at play in the leptin-driven induction of ELF3. Moreover, we confirm the participation of NFΚB in the leptin/IL-1ß synergistic induction of ELF3. CONCLUSION: Here we show, for the first time, the regulation of ELF3 expression by leptin, suggesting that this transcription factor likely mediates the inflammatory responses triggered by leptin in articular chondrocytes.


Subject(s)
Chondrocytes/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Inflammation/genetics , Leptin/immunology , Obesity/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Cartilage/immunology , Cartilage/metabolism , Cell Line , Cells, Cultured , Chondrocytes/immunology , DNA-Binding Proteins/immunology , Humans , Inflammation/immunology , Interleukin-1beta/immunology , Leptin/genetics , Obesity/immunology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/immunology , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Leptin/genetics , Receptors, Leptin/immunology , Transcription Factors/immunology , Transcriptional Activation
7.
Front Immunol ; 9: 497, 2018.
Article in English | MEDLINE | ID: mdl-29593737

ABSTRACT

This study was designed to define gene expression and H3K4me3 histone modifications in T cells, B cells, and monocytes in systemic lupus erythematosus (SLE). Array studies of total peripheral blood mononuclear cells have demonstrated gene expression signatures related to neutrophils, interferon, and other inflammatory pathways. It is not clear how consistent these effects are across different cell types. In this study, RNA-seq and chromatin immunoprecipitation-seq were utilized to identify gene expression patterns and H3K4me3 histone modifications related to promoter activation in SLE. Across the three cell types, there was 55% concordance for gene expression changes related to SLE. Key conserved pathways were ribosome biogenesis among upregulated genes and heat shock response among downregulated genes. ETS family transcription factors (TFs) and STAT1 were revealed as common regulators by position weight matrices. When epigenetic changes were leveraged with gene expression, the pivotal TFs ATF3 and FOS were defined with ATF3 also cross-referencing with gene expression-identified TFs. Genome-wide association study (GWAS) single nucleotide polymorphisms associated with SLE were cross-referenced with both mRNA and H3K4me3 changes in SLE. Baseline mRNA expression and H3K4me3 peak height was higher at sites that cross-referenced with GWAS signals, however, all three cell types exhibited an overall decrease in expression of GWAS-associated RNAs differentially expressed in SLE. H3K4me3 changes in SLE were also enriched in GWAS-associated sites. In summary, the SLE disease process is associated with both shared and cell-specific changes in gene expression and epigenetics. Surprisingly, GWAS-associated RNAs were overall markedly decreased across all three cell types. TF analysis identified ATF3, FOS, STAT1, and ETS family members as critical, all pathways with a recognized relationship to the SLE disease process. GWAS signals clearly mark both cell-type specific changes in SLE as well as concordant changes across all three cell types. Interpretation of single nucleotide polymorphism effects in SLE will require tissue-specific mechanistic studies and therapeutics will require mechanistic studies in multiple cell types.


Subject(s)
Down-Regulation/immunology , Histones/immunology , Lupus Erythematosus, Systemic/immunology , RNA, Messenger/immunology , Signal Transduction/immunology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/immunology , Female , Genome-Wide Association Study , Histones/genetics , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , RNA, Messenger/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Signal Transduction/genetics
8.
Nat Commun ; 8(1): 1426, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127283

ABSTRACT

Humoral immunity requires B cells to respond to multiple stimuli, including antigen, membrane and soluble ligands, and microbial products. Ets family transcription factors regulate many aspects of haematopoiesis, although their functions in humoral immunity are difficult to decipher as a result of redundancy between the family members. Here we show that mice lacking both PU.1 and SpiB in mature B cells do not generate germinal centers and high-affinity antibody after protein immunization. PU.1 and SpiB double-deficient B cells have a survival defect after engagement of CD40 or Toll-like receptors (TLR), despite paradoxically enhanced plasma cell differentiation. PU.1 and SpiB regulate the expression of many components of the B cell receptor signaling pathway and the receptors for CD40L, BAFF and TLR ligands. Thus, PU.1 and SpiB enable B cells to appropriately respond to environmental cues.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Animals , B-Lymphocytes/cytology , CD40 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets/deficiency , Proto-Oncogene Proteins c-ets/genetics , Signal Transduction , Toll-Like Receptors/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics
9.
Cell Mol Biol (Noisy-le-grand) ; 63(8): 19-22, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28886309

ABSTRACT

Despite advances in treatment, children with acute lymphoblastic leukemia (ALL) still experience drug resistance and relapse. Several gene mutations are involved in the onset of this disease and resistance to therapy. The present study examines the incidence of IKZF1, CDKN2A/B, PAX5, EBF1, ETV6, BTG1, RB1, JAK2, and Xp22.33 gene deletions/duplications associated with pediatric ALL in Iran and investigates the possible effect of these mutations on drug resistance. Three-year disease-free survival (3DFS) was evaluated for children diagnosed with Philadelphia negative precursor-B-cell ALL hospitalized at Sayed-al-Shohada Hospital, Isfahan-Iran, from January 2009 until December 2012. DNA was extracted from bone marrow slides, and ALL correlated gene deletions and duplications were measured using Multiplex Ligation-dependent Probe Amplification (MLPA) method. The correlation between gene mutations and 3DFS was then assessed. Among the nine aforementioned investigated genes, 63% of samples showed at least one gene mutation. At least two concomitant genomic mutations were observed in 42% of samples. Pax5 deletion was the most prevalent gene mutation observed in 45% of cases, and showed significant negative impact on response to treatment. CDKN2A/B (9p21.3) gene deletion, and ETV6 (12p13.2) gene duplication also demonstrated negative effect on patient survival and contributed to a worse prognosis if concomitant with Pax5 gene deletion. ALL patients with one of the gene deletions including Pax5  and CDKN2A/B (9p21.3) or ETV6 (12p13.2) gene duplication are classified as high-risk patients and need more intensified protocols of treatment to improve their chance of survival.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Gene Deletion , Gene Expression Regulation, Leukemic , PAX5 Transcription Factor/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Adolescent , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p15/immunology , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p18/immunology , Drug Resistance, Neoplasm/genetics , Female , Gene Duplication , Humans , Infant , Iran , Male , PAX5 Transcription Factor/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , Prognosis , Proto-Oncogene Proteins c-ets/immunology , Repressor Proteins/immunology , Survival Analysis , ETS Translocation Variant 6 Protein
10.
Sci Signal ; 10(475)2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28420758

ABSTRACT

Goblet cell metaplasia and excessive mucus secretion associated with asthma, cystic fibrosis, and chronic obstructive pulmonary disease contribute to morbidity and mortality worldwide. We performed a high-throughput screen to identify small molecules targeting a transcriptional network critical for the differentiation of goblet cells in response to allergens. We identified RCM-1, a nontoxic small molecule that inhibited goblet cell metaplasia and excessive mucus production in mice after exposure to allergens. RCM-1 blocked the nuclear localization and increased the proteasomal degradation of Forkhead box M1 (FOXM1), a transcription factor critical for the differentiation of goblet cells from airway progenitor cells. RCM-1 reduced airway resistance, increased lung compliance, and decreased proinflammatory cytokine production in mice exposed to the house dust mite and interleukin-13 (IL-13), which triggers goblet cell metaplasia. In cultured airway epithelial cells and in mice, RCM-1 reduced IL-13 and STAT6 (signal transducer and activator of transcription 6) signaling and prevented the expression of the STAT6 target genes Spdef and Foxa3, which are key transcriptional regulators of goblet cell differentiation. These results suggest that RCM-1 is an inhibitor of goblet cell metaplasia and IL-13 signaling, providing a new therapeutic candidate to treat patients with asthma and other chronic airway diseases.


Subject(s)
Allergens/toxicity , Forkhead Box Protein M1/antagonists & inhibitors , Goblet Cells/immunology , Interleukin-13/immunology , STAT6 Transcription Factor/immunology , Animals , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/immunology , Goblet Cells/pathology , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/immunology , Interleukin-13/genetics , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/immunology , Metaplasia/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , STAT6 Transcription Factor/genetics
11.
J Leukoc Biol ; 101(5): 1109-1117, 2017 05.
Article in English | MEDLINE | ID: mdl-28235774

ABSTRACT

Megakaryocytes (MK) are the sole source of platelets in the body. They develop from lineage-committed hematopoietic stem and progenitor cells (HSPCs) via intermediate cells, which differ in morphology, size, ploidy, and surface phenotype. Development and maturation of MKs is governed by different transcription factors, including GATA-1, E26 transformation-specific transcription factor (ETS) family members, nuclear factor erythroid 2 transcription factor (NF-E2), and STAT3. During such challenges as acute inflammation, platelets are consumed in high numbers and must be replenished to secure survival of the host. This is achieved by integration of inflammatory signals into early MK development and depends on the STAT1-mediated enhanced translation of transcripts in stem cell-like megakaryocyte progenitors. Here, we review recent developments, which highlight the impact of inflammation on the development of platelets from HSPCs.


Subject(s)
Blood Platelets/immunology , Cytokines/immunology , Gene Expression Regulation, Developmental/immunology , Hematopoietic Stem Cells/immunology , Inflammation/immunology , Megakaryocytes/immunology , Animals , Blood Platelets/cytology , Cell Differentiation , Cell Lineage/immunology , Cell Proliferation , Cytokines/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/immunology , Hematopoietic Stem Cells/cytology , Humans , Inflammation/genetics , Inflammation/pathology , Megakaryocytes/cytology , Mice , NF-E2 Transcription Factor, p45 Subunit/genetics , NF-E2 Transcription Factor, p45 Subunit/immunology , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , Signal Transduction
12.
Blood ; 129(4): 509-519, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27940477

ABSTRACT

Macrophages are key components of the innate immune system and play pivotal roles in immune response, organ development, and tissue homeostasis. Studies in mice and zebrafish have shown that tissue-resident macrophages derived from different hematopoietic origins manifest distinct developmental kinetics and colonization potential, yet the genetic programs controlling the development of macrophages of different origins remain incompletely defined. In this study, we use zebrafish, where tissue-resident macrophages arise from the rostral blood island (RBI) and ventral wall of dorsal aorta (VDA), the zebrafish hematopoietic tissue equivalents to the mouse yolk sac and aorta-gonad-mesonephros for myelopoiesis, to address this issue. We show that RBI- and VDA-born macrophages are orchestrated by distinctive regulatory networks formed by the E-twenty-six (Ets) transcription factors Pu.1 and Spi-b, the zebrafish ortholog of mouse spleen focus forming virus proviral integration oncogene B (SPI-B), and the helix-turn-helix DNA-binding domain containing protein Irf8. Epistatic studies document that during RBI macrophage development, Pu.1 acts upstream of Spi-b, which, upon induction by Pu.1, partially compensates the function of Pu.1. In contrast, Pu.1 and Spi-b act in parallel and cooperatively to regulate the development of VDA-derived macrophages. Interestingly, these two distinct regulatory networks orchestrate the RBI- and VDA-born macrophage development largely by regulating a common downstream gene, Irf8. Our study indicates that macrophages derived from different origins are governed by distinct genetic networks formed by the same repertoire of myeloid-specific transcription factors.


Subject(s)
Cell Lineage/immunology , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Zebrafish/immunology , Amino Acid Sequence , Animals , Aorta/cytology , Aorta/growth & development , Aorta/immunology , Cell Differentiation , Cell Lineage/genetics , Embryo, Nonmammalian , Humans , Immunity, Innate , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Macrophages/cytology , Mesonephros/cytology , Mesonephros/growth & development , Mesonephros/immunology , Mice , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , Signal Transduction , Trans-Activators/genetics , Yolk Sac/cytology , Yolk Sac/growth & development , Yolk Sac/immunology , Zebrafish/genetics , Zebrafish/growth & development
13.
Oncotarget ; 7(11): 12191-205, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26919255

ABSTRACT

The t(12;21) translocation generating the ETV6/RUNX1 fusion gene represents the most frequent chromosomal rearrangement in childhood leukemia. Presence of ETV6/RUNX1 alone is usually not sufficient for leukemia onset, and additional genetic alterations have to occur in ETV6/RUNX1-positive cells to cause transformation. We have previously generated an ETV6/RUNX1 transgenic mouse model where the expression of the fusion gene is restricted to CD19-positive B cells. Since BCL2 family members have been proposed to play a role in leukemogenesis, we investigated combined effects of ETV6/RUNX1 with exogenous expression of the antiapoptotic protein BCL2 by crossing ETV6/RUNX1 transgenic animals with Vav-BCL2 transgenic mice. Strikingly, co-expression of ETV6/RUNX1 and BCL2 resulted in significantly shorter disease latency in mice, indicating oncogene cooperativity. This was associated with faster development of follicular B cell lymphoma and exacerbated immune complex glomerulonephritis. ETV6/RUNX1-BCL2 double transgenic animals displayed increased B cell numbers and immunoglobulin titers compared to Vav-BCL2 transgenic mice. This led to pronounced deposition of immune complexes in glomeruli followed by accelerated development of immune complex glomerulonephritis. Thus, our study reveals a previously unrecognized synergism between ETV6/RUNX1 and BCL2 impacting on malignant disease and autoimmunity.


Subject(s)
Core Binding Factor Alpha 2 Subunit/immunology , Glomerulonephritis/immunology , Immunoglobulins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/immunology , Proto-Oncogene Proteins c-ets/immunology , Repressor Proteins/immunology , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Glomerulonephritis/genetics , Humans , Immunoglobulins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , ETS Translocation Variant 6 Protein
14.
Mol Cell Biol ; 35(9): 1619-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25733685

ABSTRACT

Generation of antibodies against T-independent and T-dependent antigens requires Toll-like receptor (TLR) engagement on B cells for efficient responses. However, the regulation of TLR expression and responses in B cells is not well understood. PU.1 and Spi-B (encoded by Sfpi1 and Spib, respectively) are transcription factors of the E26 transformation-specific (ETS) family and are important for B cell development and function. It was found that B cells from mice knocked out for Spi-B and heterozygous for PU.1 (Sfpi1(+/-) Spib(-/-) [PUB] mice) proliferated poorly in response to TLR ligands compared to wild-type (WT) B cells. The NF-κB family member p50 (encoded by Nfkb1) is required for lipopolysaccharide (LPS) responsiveness in mice. PUB B cells expressed reduced Nfkb1 mRNA transcripts and p50 protein. The Nfkb1 promoter was regulated directly by PU.1 and Spi-B, as shown by reporter assays and chromatin immunoprecipitation analysis. Occupancy of the Nfkb1 promoter by PU.1 was reduced in PUB B cells compared to that in WT B cells. Finally, infection of PUB B cells with a retroviral vector encoding p50 substantially restored proliferation in response to LPS. We conclude that Nfkb1 transcriptional activation by PU.1 and Spi-B promotes TLR-mediated B cell proliferation.


Subject(s)
B-Lymphocytes/cytology , NF-kappa B p50 Subunit/immunology , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins/immunology , Spleen/cytology , Toll-Like Receptors/immunology , Trans-Activators/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cell Proliferation , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/genetics , RNA, Messenger/genetics , Transcriptional Activation
15.
J Immunol ; 194(8): 3798-807, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25769919

ABSTRACT

Spi-C is an E26 transformation-specific family transcription factor that is highly related to PU.1 and Spi-B. Spi-C is expressed in developing B cells, but its function in B cell development and function is not well characterized. To determine whether Spi-C functions as a negative regulator of Spi-B (encoded by Spib), mice were generated that were germline knockout for Spib and heterozygous for Spic (Spib(-/-)Spic(+/-)). Interestingly, loss of one Spic allele substantially rescued B cell frequencies and absolute numbers in Spib(-/-) mouse spleens. Spib(-/-)Spic(+/-) B cells had restored proliferation compared with Spib(-/-) B cells in response to anti-IgM or LPS stimulation. Investigation of a potential mechanism for the Spib(-/-)Spic(+/-) phenotype revealed that steady-state levels of Nfkb1, encoding p50, were elevated in Spib(-/-)Spic(+/-) B cells compared with Spib(-/-) B cells. Spi-B was shown to directly activate the Nfkb1 gene, whereas Spi-C was shown to repress this gene. These results indicate a novel role for Spi-C as a negative regulator of B cell development and function.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , DNA-Binding Proteins/immunology , Gene Expression Regulation/immunology , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/immunology , Spleen/immunology
16.
Clin Exp Allergy ; 45(9): 1447-58, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25772331

ABSTRACT

BACKGROUND: Increased mucus production is a critical factor impairing lung function in patients suffering from bronchial asthma, the most common chronic inflammatory lung disease worldwide. OBJECTIVE: This study aimed at investigating whether goblet cell (GC) metaplasia and mucus production are differentially regulated in proximal and distal airways. METHODS: Female Balb/c mice were sensitized to ovalbumin (OVA) and challenged with an OVA-aerosol on two consecutive days for 1 week (acute) or 12 weeks (chronic). Real-time RT-PCR analysis was applied on microdissected airways. RESULTS: In acutely and chronically OVA-challenged mice, GC metaplasia and mucus production were observed in proximal but not in distal airways. In contrast, inflammation reflected by the infiltration of eosinophils and expression of the TH2-type cytokines IL-4 and IL-13 was increased in both proximal and distal airways. Abundance of IL-13Rα1 was lower in distal airways of healthy control mice. Under acute and chronic OVA-exposure, activation of IL-13Rα1-dependent signalling cascade, reflected by Spdef and Foxo3A transcription factors, was attenuated in distal compared to proximal airways. CONCLUSION AND CLINICAL RELEVANCE: These data indicate that distal airways might be less sensitive to IL-13-induced GC metaplasia and mucus production through lower expression of IL-13Rα1 and attenuated activation of downstream signalling. This might represent a protective strategy to prevent mucus plugging of distal airways and thus impaired ventilation of attached alveoli.


Subject(s)
Asthma/immunology , Gene Expression Regulation/immunology , Goblet Cells/immunology , Interleukin-13/immunology , Lung/immunology , Signal Transduction/immunology , Animals , Asthma/metabolism , Asthma/pathology , Female , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Goblet Cells/metabolism , Goblet Cells/pathology , Interleukin-13/biosynthesis , Interleukin-13 Receptor alpha1 Subunit/biosynthesis , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Lung/metabolism , Lung/pathology , Metaplasia , Mice , Mice, Inbred BALB C , Mucus/immunology , Mucus/metabolism , Proto-Oncogene Proteins c-ets/biosynthesis , Proto-Oncogene Proteins c-ets/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
17.
J Leukoc Biol ; 97(1): 171-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25351511

ABSTRACT

ETS family proteins play a role in immune responses. A unique member of this family, Elk-3, is a transcriptional repressor that regulates the expression of HO-1. Elk-3 is very sensitive to the effects of inflammatory mediators and is down-regulated by bacterial endotoxin (LPS). In the present study, exposure of mouse macrophages to Escherichia coli LPS resulted in decreased, full-length, and splice-variant isoforms of Elk-3. We isolated the Elk-3 promoter and demonstrated that LPS also decreased promoter activity. The Elk-3 promoter contains GC-rich regions that are putative binding sites for zinc-finger transcription factors, such as Sp1 and KLFs. Mutation of the GC-rich region from bp -613 to -603 blunted LPS-induced down-regulation of the Elk-3 promoter. Similar to the LPS response, coexpression of KLF4 led to repression of Elk-3 promoter activity, whereas coexpression of Sp1 increased activity. ChIP assays revealed that KLF4 binding to the Elk-3 promoter was increased by LPS exposure, and Sp1 binding was decreased. Thus, down-regulation of Elk-3 by bacterial LPS is regulated, in part, by the transcriptional repressor KLF4. Overexpression of Elk-3, in the presence of E. coli bacteria, resulted in decreased macrophage phagocytosis. To determine whether limited expression of HO-1 may contribute to this response, we exposed HO-1-deficient bone marrow-derived macrophages to E. coli and found a comparable reduction in bacterial phagocytosis. These data suggest that down-regulation of Elk-3 and the subsequent induction of HO-1 are important for macrophage function during the inflammatory response to infection.


Subject(s)
Gene Expression Regulation/immunology , Kruppel-Like Transcription Factors/immunology , Macrophages/immunology , Phagocytosis/immunology , Proto-Oncogene Proteins c-ets/immunology , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Escherichia coli Infections/immunology , Heme Oxygenase-1/immunology , Inflammation/immunology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phagocytosis/genetics , Proto-Oncogene Proteins c-ets/genetics , Real-Time Polymerase Chain Reaction , Transfection
18.
J Exp Med ; 211(12): 2425-38, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25385757

ABSTRACT

Medullary thymic epithelial cells (mTECs) expressing the autoimmune regulator AIRE and various tissue-specific antigens (TSAs) are critical for preventing the onset of autoimmunity and may attenuate tumor immunity. However, molecular mechanisms controlling mTEC development remain elusive. Here, we describe the roles of the transcription factor Spi-B in mTEC development. Spi-B is rapidly up-regulated by receptor activator of NF-κB ligand (RANKL) cytokine signaling, which triggers mTEC differentiation, and in turn up-regulates CD80, CD86, some TSAs, and the natural inhibitor of RANKL signaling, osteoprotegerin (OPG). Spi-B-mediated OPG expression limits mTEC development in neonates but not in embryos, suggesting developmental stage-specific negative feedback regulation. OPG-mediated negative regulation attenuates cellularity of thymic regulatory T cells and tumor development in vivo. Hence, these data suggest that this negative RANKL-Spi-B-OPG feedback mechanism finely tunes mTEC development and function and may optimize the trade-off between prevention of autoimmunity and induction of antitumor immunity.


Subject(s)
Epithelial Cells/immunology , Immune Tolerance/immunology , Proto-Oncogene Proteins c-ets/immunology , Thymus Gland/immunology , Animals , Animals, Newborn , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Epithelial Cells/metabolism , Feedback, Physiological , Female , Gene Expression/immunology , Immune Tolerance/genetics , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/immunology , Osteoprotegerin/metabolism , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RANK Ligand/immunology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/immunology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Thymus Gland/metabolism , NF-kappaB-Inducing Kinase
19.
J Allergy Clin Immunol ; 127(1): 254-61, 261.e1-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21126757

ABSTRACT

BACKGROUND: Asthma is a major public health burden worldwide. Studies from our group and others have demonstrated that SERPINB3 and SERPINB4 are induced in patients with asthma; however, their mechanistic role in asthma has yet to be determined. OBJECTIVE: To evaluate the role of Serpin3a, the murine homolog of SERPINB3 and SERPINB4, in asthma. METHODS: We studied wild-type Balb/c and Serpinb3a-null mice in house dust mite or IL-13-induced asthma models and evaluated airway hyperresponsiveness, inflammation, and goblet cell hyperplasia. RESULTS: Airway hyperresponsiveness and goblet cell hyperplasia were markedly attenuated in the Serpinb3a-null mice compared with the wild-type mice after allergen challenge, with minimal effects on inflammation. Expression of sterile alpha motif pointed domain containing v-ets avian erythroblastosis virus E26 oncogene homolog transcription factor (SPDEF), a transcription factor that mediates goblet cell hyperplasia, was decreased in the absence of Serpinb3a. IL-13-treated Serpinb3a-null mice showed attenuated airway hyperresponsiveness, inflammation, and mucus production. CONCLUSION: Excessive mucus production and mucus plugging are key pathologic features of asthma, yet the mechanisms responsible for mucus production are not well understood. Our data reveal a novel nonredundant role for Serpinb3a in mediating mucus production through regulation of SPDEF expression. This pathway may be used to target mucus hypersecretion effectively.


Subject(s)
Asthma/immunology , Mucus/immunology , Proto-Oncogene Proteins c-ets/immunology , Serpins/immunology , Animals , Asthma/metabolism , Asthma/pathology , Bronchoalveolar Lavage Fluid , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Gene Expression Regulation/immunology , Goblet Cells/immunology , Goblet Cells/metabolism , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucus/metabolism , Proto-Oncogene Proteins c-ets/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Serpins/metabolism
20.
J Immunol ; 185(12): 7374-84, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21057087

ABSTRACT

Splenic B-2 cells can be divided into two major subsets: follicular (FO) and marginal zone (MZ) B cells. FO and MZ B cells are generated from immature transitional B cells. Few transcription factors have been identified that regulate FO B cell differentiation. The highly related proteins PU.1, Spi-B, and Spi-C are transcription factors of the E26-transformation-specific family and are important for B cell differentiation and function. To determine whether these proteins play a role in the differentiation of FO B cells, we performed a detailed analysis of splenic B cells in mice with inactivating mutations in the genes encoding PU.1 (Sfpi1) or Spi-B (Spib). Sfpi1(+/-) Spib(-/-) (PUB) mice had a 9-fold reduction in the frequency of CD23(+) FO B cells compared with that of wild-type mice. In contrast, PUB mice had a 2-fold increase in the frequency of MZ B cells that was confirmed by immunofluorescence staining. Expression of Spi-C in Eµ-Spi-C transgenic PUB mice partially rescued frequencies of CD23(+) B cells. Gene expression analysis, in vitro reporter assays, and chromatin immunoprecipitation experiments showed that transcription of the Fcer2a gene encoding CD23 is activated by PU.1, Spi-B, and Spi-C. These results demonstrate that FO B cell differentiation is regulated by the E26-transformation-specific transcription factors PU.1, Spi-B, and Spi-C.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins/immunology , Proto-Oncogene Proteins c-ets/immunology , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Animals , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgE/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...