1
|
Wang S, Bao N, Li M, Liu D, Tao L. Ets2 Exacerbates Diabetic Retinopathy by Aggravating the Proliferation of Endothelial Cells and Inflammatory Response. Biochem Genet 2024:10.1007/s10528-024-10938-8. [PMID: 39432129 DOI: 10.1007/s10528-024-10938-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024]
Abstract
Proliferative diabetic retinopathy (PDR), the most common type of diabetic retinopathy, is a main cause of visual and impairment blindness. Abnormal neovascularization, endothelial dysfunction, and vascular inflammation are important mechanisms for the development of PDR. Ets2 regulates angiogenesis-related genes and inflammation, however, the effect of Ets2 in PDR procession has not been clarified. Thus, this study is performed to investigate whether Ets2 exerts key functions in PDR. In this study, 10-week-old mice were used for establishing STZ-induced diabetic mice, and Ets2 expression was analyzed in retina tissues. Besides, newborn mice were applied to construct oxygen-induced retinopathy (OIR) models. The Ets2 expression, oxidative stress, and inflammation were detected in retina tissues. We found that Ets2 was highly expressed in retina tissues both in diabetic mice and OIR mice. Oxidative stress and inflammatory processes are two factors contributing to the pathogenesis of PDR. In retinal tissues of OIR mice, Ets2 knockdown inhibited expression of inflammatory mediators VEGFA, IL-6, and IL-8, and biomarkers of oxidative stress MCP-1, VCAM-1, and iNOS. ROS production was also inhibited by silencing Ets2. Ets2 deficiency inhibited endothelial cell proliferation in the retina. Furthermore, Ets2 knockdown contributed to suppressing the expression of angiogenesis-related genes VEGFA, JUNB, MMP-9, Tie2, Ang-2, and EphB4. Our study highlights that Ets2 accelerates PDR procession by promoting the proliferation of endothelial cells, oxidative stress, and inflammation, which provides a novel target against PDR.
Collapse
Affiliation(s)
- Song Wang
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Ning Bao
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Mohan Li
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Dongwei Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Liming Tao
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China.
| |
Collapse
|
2
|
Fatema K, Haidar Z, Tanim MTH, Nath SD, Sajib AA. Unveiling the link between arsenic toxicity and diabetes: an in silico exploration into the role of transcription factors. Toxicol Res 2024; 40:653-672. [PMID: 39345741 PMCID: PMC11436564 DOI: 10.1007/s43188-024-00255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024] Open
Abstract
Arsenic-induced diabetes, despite being a relatively newer finding, is now a growing area of interest, owing to its multifaceted nature of development and the diversity of metabolic conditions that result from it, on top of the already complicated manifestation of arsenic toxicity. Identification and characterization of the common and differentially affected cellular metabolic pathways and their regulatory components among various arsenic and diabetes-associated complications may aid in understanding the core molecular mechanism of arsenic-induced diabetes. This study, therefore, explores the effects of arsenic on human cell lines through 14 transcriptomic datasets containing 160 individual samples using in silico tools to take a systematic, deeper look into the pathways and genes that are being altered. Among these, we especially focused on the role of transcription factors due to their diverse and multifaceted roles in biological processes, aiming to comprehensively investigate the underlying mechanism of arsenic-induced diabetes as well as associated health risks. We present a potential mechanism heavily implying the involvement of the TGF-β/SMAD3 signaling pathway leading to cell cycle alterations and the NF-κB/TNF-α, MAPK, and Ca2+ signaling pathways underlying the pathogenesis of arsenic-induced diabetes. This study also presents novel findings by suggesting potential associations of four transcription factors (NCOA3, PHF20, TFDP1, and TFDP2) with both arsenic toxicity and diabetes; five transcription factors (E2F5, ETS2, EGR1, JDP2, and TFE3) with arsenic toxicity; and one transcription factor (GATA2) with diabetes. The novel association of the transcription factors and proposed mechanism in this study may serve as a take-off point for more experimental evidence needed to understand the in vivo cellular-level diabetogenic effects of arsenic. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00255-y.
Collapse
Affiliation(s)
- Kaniz Fatema
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Zinia Haidar
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Md Tamzid Hossain Tanim
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Sudipta Deb Nath
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| |
Collapse
|
3
|
Tousinas G, Emmanuel AO, Tracy M, Arnovitz S, Friedman D, Papamarcaki T, Gounari F. Stabilization of β-Catenin Directs HEB to Limit Thymic Selection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:641-650. [PMID: 38958395 PMCID: PMC11333165 DOI: 10.4049/jimmunol.2400160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Activation of β-catenin in CD4+CD8+ double-positive (DP) thymocytes halts development before the thymic selection stage and predisposes to transformation. Leukemogenesis, but not the developmental block, depends on TCF-1, β-catenin's DNA-binding partner. In this study, we show that β-catenin activation directs the DNA-binding protein HEB to block DP thymocyte development. Conditional loss of HEB in DP thymocytes with stabilized β-catenin restores the frequencies of postselection TCRβhi/CCR7+ and TCRβhi/CD69+ DPs and their cell-cycle profile. This recovery is associated with significant reversal of β-catenin-induced expression changes, particularly those related to the CD69+ DP cell signature and to cell-cycle pathways. Stabilizing β-catenin in DP thymocytes directs HEB binding to ≈11,000 novel DNA sites throughout the genome. Novel HEB sites mark most CD69+ DP cell signature genes that change expression upon activation of β-catenin and then revert after loss of HEB. Moreover, many of the novel HEB sites occupy promoter regions of genes enriched in mitotic cell cycle pathways. HEB binding to those regions correlates with downregulation of the associated genes, and HEB inactivation restores expression to physiologic levels. These findings highlight a molecular interplay between HEB and β-catenin that can impair thymic development.
Collapse
Affiliation(s)
- Georgios Tousinas
- Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
| | | | - Melissa Tracy
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Stephen Arnovitz
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | - David Friedman
- Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259
| | - Thomais Papamarcaki
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10 Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 451 10 Ioannina, Greece
| | - Fotini Gounari
- Department of Immunology, Mayo Clinic, Scottsdale, AZ 85259
| |
Collapse
|
4
|
Qin Y, Pu X, Hu D, Yang M. Machine learning-based biomarker screening for acute myeloid leukemia prognosis and therapy from diverse cell-death patterns. Sci Rep 2024; 14:17874. [PMID: 39090256 PMCID: PMC11294352 DOI: 10.1038/s41598-024-68755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Acute myeloid leukemia (AML) exhibits pronounced heterogeneity and chemotherapy resistance. Aberrant programmed cell death (PCD) implicated in AML pathogenesis suggests PCD-related signatures could serve as biomarkers to predict clinical outcomes and drug response. We utilized 13 PCD pathways, including apoptosis, pyroptosis, ferroptosis, autophagy, necroptosis, cuproptosis, parthanatos, entotic cell death, netotic cell death, lysosome-dependent cell death, alkaliptosis, oxeiptosis, and disulfidptosis to develop predictive models based on 73 machine learning combinations from 10 algorithms. Bulk RNA-sequencing, single-cell RNA-sequencing transcriptomic data, and matched clinicopathological information were obtained from the TCGA-AML, Tyner, and GSE37642-GPL96 cohorts. These datasets were leveraged to construct and validate the models. Additionally, in vitro experiments were conducted to substantiate the bioinformatics findings. The machine learning approach established a 6-gene pan-programmed cell death-related genes index (PPCDI) signature. Validation in two external cohorts showed high PPCDI associated with worse prognosis in AML patients. Incorporating PPCDI with clinical variables, we constructed several robust prognostic nomograms that accurately predicted prognosis of AML patients. Multi-omics analysis integrating bulk and single-cell transcriptomics revealed correlations between PPCDI and immunological features, delineating the immune microenvironment landscape in AML. Patients with high PPCDI exhibited resistance to conventional chemotherapy like doxorubicin but retained sensitivity to dasatinib and methotrexate (FDA-approved drugs for other leukemias), suggesting the potential of PPCDI to guide personalized therapy selection in AML. In summary, we developed a novel PPCDI model through comprehensive analysis of diverse programmed cell death pathways. This PPCDI signature demonstrates great potential in predicting clinical prognosis and drug sensitivity phenotypes in AML patients.
Collapse
Affiliation(s)
- Yu Qin
- Department of Hematology, First Affiliated Hospital of Anhui Medical University, 218Jixi Road, Hefei, 230022, Anhui, China
| | - Xuexue Pu
- Department of Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, 218Jixi Road, Hefei, 230022, Anhui, China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai, China
| | - Mingzhen Yang
- Department of Hematology, First Affiliated Hospital of Anhui Medical University, 218Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
5
|
Thompson B, Strange A, Amato CM, Hester-McCullough J, Sarnaik AA, Weber JS, Woods DM. CD4 Phenotypes Are Associated with Reduced Expansion of Tumor-Infiltrating Lymphocytes in Melanoma Patients Treated with Adoptive Cell Therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:735-742. [PMID: 37466381 PMCID: PMC10528290 DOI: 10.4049/jimmunol.2300250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/04/2023] [Indexed: 07/20/2023]
Abstract
Tumor-infiltrating lymphocyte (TIL) adoptive cell therapy is effective in treating malignant melanoma, but its success relies on the adequate ex vivo expansion of TIL. To assess correlates of TIL expansion, CD4+ and CD8+ TIL were analyzed by RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing of acetylated histone 3. Patients were grouped into "TIL high" and "TIL low" based on division at the median number of TIL infused. Greater numbers of TIL infused correlated with longer overall survival, and increased frequencies of CD4+ cells infused were negatively correlated with the number of TIL infused. RNA-seq analysis of CD4+ TIL showed increases in Th2/Th17/regulatory T cell-related transcripts and pathways in the TIL-low group. Analysis of a public single-cell RNA-seq dataset validated findings that increased frequencies of CD4+ cells were negatively correlated with the number of TIL infused. TIL-low patients had significantly increased frequencies of CD4+ cells expressing ETS2 and OSM and trended toward increased expression of TNFRSF18.
Collapse
Affiliation(s)
- Brian Thompson
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO
| | - Ann Strange
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO
| | - Carol M. Amato
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO
| | | | - Amod A. Sarnaik
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jeffrey S. Weber
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - David M. Woods
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO
| |
Collapse
|
6
|
Schwarzfischer M, Niechcial A, Handler K, Morsy Y, Wawrzyniak M, Laimbacher AS, Atrott K, Manzini R, Baebler K, Hering L, Katkeviciutė E, Häfliger J, Lang S, Keller ME, Woodtli J, Eisenbeiss L, Kraemer T, Schraner EM, Wiesendanger M, Zeissig S, Rogler G, Moor AE, Scharl M, Spalinger MR. TiO 2 nanoparticles abrogate the protective effect of the Crohn's disease-associated variation within the PTPN22 gene locus. Gut 2022; 72:1101-1114. [PMID: 36191962 DOI: 10.1136/gutjnl-2021-325911] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/04/2022] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Inflammatory bowel disease (IBD) is a multifactorial condition driven by genetic and environmental risk factors. A genetic variation in the protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene has been associated with autoimmune disorders while protecting from the IBD subtype Crohn's disease. Mice expressing the murine orthologous PTPN22-R619W variant are protected from intestinal inflammation in the model of acute dextran sodium sulfate (DSS)-induced colitis. We previously identified food-grade titanium dioxide (TiO2, E171) as a neglected IBD risk factor. Here, we investigate the interplay of the PTPN22 variant and TiO2-mediated effects during IBD pathogenesis. DESIGN Acute DSS colitis was induced in wild-type and PTPN22 variant mice (PTPN22-R619W) and animals were treated with TiO2 nanoparticles during colitis induction. Disease-triggering mechanisms were investigated using bulk and single-cell RNA sequencing. RESULTS In mice, administration of TiO2 nanoparticles abrogated the protective effect of the variant, rendering PTPN22-R619W mice susceptible to DSS colitis. In early disease, cytotoxic CD8+ T-cells were found to be reduced in the lamina propria of PTPN22-R619W mice, an effect reversed by TiO2 administration. Normalisation of T-cell populations correlated with increased Ifng expression and, at a later stage of disease, the promoted prevalence of proinflammatory macrophages that triggered severe intestinal inflammation. CONCLUSION Our findings indicate that the consumption of TiO2 nanoparticles might have adverse effects on the gastrointestinal health of individuals carrying the PTPN22 variant. This demonstrates that environmental factors interact with genetic risk variants and can reverse a protective mechanism into a disease-promoting effect.
Collapse
Affiliation(s)
- Marlene Schwarzfischer
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Niechcial
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kristina Handler
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Yasser Morsy
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marcin Wawrzyniak
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andrea S Laimbacher
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kirstin Atrott
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Roberto Manzini
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Katharina Baebler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Larissa Hering
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Egle Katkeviciutė
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Janine Häfliger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Silvia Lang
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Maja E Keller
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Jérôme Woodtli
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Lisa Eisenbeiss
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Thomas Kraemer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Elisabeth M Schraner
- Institutes of Veterinary Anatomy and Virology, University of Zurich, Zurich, Switzerland
| | - Mahesa Wiesendanger
- Institutes of Veterinary Anatomy and Virology, University of Zurich, Zurich, Switzerland
| | - Sebastian Zeissig
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, and Department of Medicine I, University Medical Center Dresden, Dresden, Germany
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andreas E Moor
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marianne R Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Chromatin organizer SATB1 controls the cell identity of CD4 + CD8 + double-positive thymocytes by regulating the activity of super-enhancers. Nat Commun 2022; 13:5554. [PMID: 36138028 PMCID: PMC9500044 DOI: 10.1038/s41467-022-33333-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
Abstract
CD4+ and CD8+ double-positive (DP) thymocytes play a crucial role in T cell development in the thymus. DP cells rearrange the T cell receptor gene Tcra to generate T cell receptors with TCRβ. DP cells differentiate into CD4 or CD8 single-positive (SP) thymocytes, regulatory T cells, or invariant nature kill T cells (iNKT) in response to TCR signaling. Chromatin organizer SATB1 is highly expressed in DP cells and is essential in regulating Tcra rearrangement and differentiation of DP cells. Here we explored the mechanism of SATB1 orchestrating gene expression in DP cells. Single-cell RNA sequencing shows that Satb1 deletion changes the cell identity of DP thymocytes and down-regulates genes specifically and highly expressed in DP cells. Super-enhancers regulate the expressions of DP-specific genes, and our Hi-C data show that SATB1 deficiency in thymocytes reduces super-enhancer activity by specifically decreasing interactions among super-enhancers and between super-enhancers and promoters. Our results reveal that SATB1 plays a critical role in thymocyte development to promote the establishment of DP cell identity by globally regulating super-enhancers of DP cells at the chromatin architectural level.
Collapse
|
8
|
Davoulou P, Aggeletopoulou I, Panagoulias I, Georgakopoulos T, Mouzaki A. Transcription factor Ets-2 regulates the expression of key lymphotropic factors. Mol Biol Rep 2020; 47:7871-7881. [PMID: 33006713 DOI: 10.1007/s11033-020-05865-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/25/2020] [Indexed: 01/20/2023]
Abstract
Transcription factor Ets-2 downregulates the expression of cytokine genes and HIV-1 in resting T-cells. Herein, we studied whether Ets-2 regulates the expression of lymphotropic factors (LFs) NFAT2, NF-κΒ/p65, c-Jun, c-Fos, which regulate the activation/differentiation of T-cells, and kinase CDK10, which controls Ets-2 degradation and repression activity. In silico analysis revealed Ets-2 binding sites on the promoters of NFAT2, c-Jun, c-Fos. The T-cell lines Jurkat (models T-cell signaling/activation) and H938 (contains the HIV-1-LTR) were transfected with an Ets-2 overexpressing vector, in the presence/absence of mitogens. mRNA and protein levels were assessed by qPCR and Western immunoblotting, respectively. Ets-2 overexpression in unstimulated Jurkat increased NFAT2 and c-Jun mRNA/protein, c-Fos mRNA and NF-κΒ/p65 protein, and decreased CDK10 protein. In unstimulated H938, Ets-2 upregulated NFAT2, c-Jun and CDK10 mRNA/protein and NF-κΒ/p65 protein. In stimulated Jurkat, Ets-2 increased NFAT2, c-Jun and c-Fos mRNA/protein and decreased CDK10 mRNA/protein. In stimulated H938 Ets-2 increased NFAT2, c-Jun and c-Fos protein and reduced CDK10 protein levels. Furthermore, Ets-2 overexpression modulated the expression of pro- and anti-apoptotic genes in both cell lines. Ets-2 upregulates the expression of key LFs involved in the activation of cytokine genes or HIV-1 in T-cells, either through its physical interaction with gene promoters or through its involvement in signaling pathways that directly impact their expression. The effect of Ets-2 on CDK10 expression in H938 vs Jurkat cells dictates that, additionally to Ets-2 degradation, CDK10 may facilitate Ets-2 repression activity in cells carrying the HIV-1-LTR, contributing thus to the regulation of HIV latency in virus-infected T-cells.
Collapse
Affiliation(s)
- Panagiota Davoulou
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26500, Patras, Greece
| | - Ioanna Aggeletopoulou
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26500, Patras, Greece
| | - Ioannis Panagoulias
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26500, Patras, Greece
| | - Tassos Georgakopoulos
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26500, Patras, Greece
| | - Athanasia Mouzaki
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, 26500, Patras, Greece.
| |
Collapse
|
9
|
Yang L, Zhu Y, Kong D, Gong J, Yu W, Liang Y, Nie Y, Teng CB. EGF suppresses the expression of miR-124a in pancreatic β cell lines via ETS2 activation through the MEK and PI3K signaling pathways. Int J Biol Sci 2019; 15:2561-2575. [PMID: 31754329 PMCID: PMC6854373 DOI: 10.7150/ijbs.34985] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022] Open
Abstract
Diabetes mellitus is characterized by pancreatic β cell dysfunction. Previous studies have indicated that epidermal growth factor (EGF) and microRNA-124a (miR-124a) play opposite roles in insulin biosynthesis and secretion by beta cells. However, the underlying mechanisms remain poorly understood. In the present study, we demonstrated that EGF could inhibit miR-124a expression in beta cell lines through downstream signaling pathways, including mitogen-activated protein kinase kinase (MEK) and phosphatidylinositol 3-kinase (PI3K) cascades. Further, the transcription factor ETS2, a member of the ETS (E26 transformation-specific) family, was identified to be responsible for the EGF-mediated suppression of miR-124a expression, which was dependent on ETS2 phosphorylation at threonine 72. Activation of ETS2 decreased miR-124a promoter transcriptional activity through the putative conserved binding sites AGGAANA/TN in three miR-124a promoters located in different chromosomes. Of note, ETS2 played a positive role in regulating beta cell function-related genes, including miR-124a targets, Forkhead box a2 (FOXA2) and Neurogenic differentiation 1 (NEUROD1), which may have partly been through the inhibition of miR-124 expression. Knockdown and overexpression of ETS2 led to the prevention and promotion of insulin biosynthesis respectively, while barely affecting the secretion ability. These results suggest that EGF may induce the activation of ETS2 to inhibit miR-124a expression to maintain proper beta cell functions and that ETS2, as a novel regulator of insulin production, is a potential therapeutic target for diabetes mellitus treatment.
Collapse
Affiliation(s)
- Lin Yang
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Yuansen Zhu
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Delin Kong
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Jiawei Gong
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Wen Yu
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Yang Liang
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Yuzhe Nie
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Chun-Bo Teng
- College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
10
|
He Z, Zhang J, Huang Z, Du Q, Li N, Zhang Q, Chen Y, Sun Z. Sumoylation of RORγt regulates T H17 differentiation and thymocyte development. Nat Commun 2018; 9:4870. [PMID: 30451821 PMCID: PMC6242824 DOI: 10.1038/s41467-018-07203-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer’s patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity. The transcription factor RORγt is essential for the differentiation of TH17 cells, thymocyte development and lymphoid organogenesis. Here the authors show that the function of RORγt is regulated by PIAS4-mediated sumoylation that stabilize the interaction with SRC1 and KAT2A, to enhance the transcriptional activity of RORγt.
Collapse
Affiliation(s)
- Zhiheng He
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Jing Zhang
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA.,Irell & Manella Graduate School of Biological Sciences, City of Hope, Duarte, 91010, CA, USA
| | - Zhaofeng Huang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Qian Du
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Ning Li
- Department of Infectious Diseases, Huashan Hospital, Fudan Univerity, Shanghai, 200040, China
| | - Qiang Zhang
- Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| | - Yuan Chen
- Division of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA
| | - Zuoming Sun
- Division of Molecular Immunology, Beckman Research Institute of City of Hope, Duarte, 91010, CA, USA.
| |
Collapse
|
11
|
Ullah F, Bhattarai D, Cheng Z, Liang X, Deng T, Rehman ZU, Talpur HS, Worku T, Brohi RD, Safdar M, Ahmad MJ, Salim M, Khan M, Ahmad HI, Zhang S. Comparative Analysis of V-Akt Murine Thymoma Viral Oncogene Homolog 3 (AKT3) Gene between Cow and Buffalo Reveals Substantial Differences for Mastitis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1463732. [PMID: 29862252 PMCID: PMC5976927 DOI: 10.1155/2018/1463732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 12/31/2017] [Accepted: 01/14/2018] [Indexed: 11/17/2022]
Abstract
AKT3 gene is a constituent of the serine/threonine protein kinase family and plays a crucial role in synthesis of milk fats and cholesterol by regulating activity of the sterol regulatory element binding protein (SREBP). AKT3 is highly conserved in mammals and its expression levels during the lactation periods of cattle are markedly increased. AKT3 is highly expressed in the intestine followed by mammary gland and it is also expressed in immune cells. It is involved in the TLR pathways as effectively as proinflammatory cytokines. The aims of this study were to investigate the sequences differences between buffalo and cow. Our results showed that there were substantial differences between buffalo and cow in some exons and noteworthy differences of the gene size in different regions. We also identified the important consensus sequence motifs, variation in 2000 upstream of ATG, substantial difference in the "3'UTR" region, and miRNA association in the buffalo sequences compared with the cow. In addition, genetic analyses, such as gene structure, phylogenetic tree, position of different motifs, and functional domains, were performed to establish their correlation with other species. This may indicate that a buffalo breed has potential resistance to disease, environment changes, and airborne microorganisms and some good production and reproductive traits.
Collapse
Affiliation(s)
- Farman Ullah
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Faculty of Veterinary and Animal Sciences, Department of Animal Breeding and Genetics, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Balochistan, Pakistan
| | - Dinesh Bhattarai
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhangrui Cheng
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, Hatfield, Hertfordshire AL9 7TA, UK
| | - Xianwei Liang
- The Opening Project of Guangxi Key Laboratory of Buffalo Genetics, Reproduction and Breeding, Nanning, Guangxi 530000, China
| | - Tingxian Deng
- The Opening Project of Guangxi Key Laboratory of Buffalo Genetics, Reproduction and Breeding, Nanning, Guangxi 530000, China
| | - Zia Ur Rehman
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Department of Animal Health, Faculty of Animal Husbandry and Veterinary Sciences, The University of Agriculture, Peshawar, Pakistan
| | - Hira Sajjad Talpur
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tesfaye Worku
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Rahim Dad Brohi
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Safdar
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Jamil Ahmad
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Mohammad Salim
- Department of Forestry and Wildlife Management, The University of Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Momen Khan
- Livestock and Dairy Development, Khyber Pakhtunkhwa, Pakistan
| | - Hafiz Ishfaq Ahmad
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Shujun Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
12
|
Panagoulias I, Karagiannis F, Aggeletopoulou I, Georgakopoulos T, Argyropoulos CP, Akinosoglou K, Gogos C, Skoutelis A, Mouzaki A. Ets-2 Acts As a Transcriptional Repressor of the Human Immunodeficiency Virus Type 1 through Binding to a Repressor-Activator Target Sequence of 5'-LTR. Front Immunol 2018; 8:1924. [PMID: 29354130 PMCID: PMC5758550 DOI: 10.3389/fimmu.2017.01924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/15/2017] [Indexed: 01/02/2023] Open
Abstract
HIV-1 is transcriptionally active in activated T helper (Th)-cells and inactive in naive or resting memory Th-cells. Ets-2 is a preinduction transcriptional repressor of the IL-2 gene in naive Th-cells and a candidate transcriptional repressor of HIV-1 in the same cells, because the −279 to −250 upstream region of HIV-1-LTR [repressor–activator target sequence (RATS)], that participates in HIV-1-LTR transcriptional silencing, encompasses the AAGGAG Ets-2 binding site. In this proof of concept study, we investigated whether Ets-2 represses the expression of HIV-1. To assess whether Ets-2 can repress HIV-1 transcriptional activation acting through RATS, we transfected Jurkat cells with an Ets-2 overexpression plasmid (pCDNA3-ets-2) or Ets-2 silencing plasmids (ets-2-shRNA) and, as target genes, plasmids carrying the whole HIV-1-LTR sequence (HIV-1-LTR-CAT) or two copies of the RATS sequence (2× RATS-CAT) or a point mutation in the Ets-2 binding site (2× mutantRATS-CAT) or CMV-CAT (control). Ets-2 overexpression resulted in a significant reduction of HIV-1-LTR-CAT and 2× RATS-CAT activities in stimulated cells, but not of the 2× mutantRATS-CAT or CMV-CAT. Ets-2 silencing led to increased activities of HIV-1-LTR-CAT and 2× RATS-CAT in unstimulated cells, but had no effect on the activities of 2× mutantRATS-CAT and CMV-CAT. To assess Ets-2 binding to HIV-1-LTR–RATS in naive Th-cells, we isolated naive Th-cell nuclear proteins and passed them through an Ets-2 antibody column; electrophoretic mobility shift assays were performed using an RATS probe mixed with consecutive protein eluates. Ets-2 bound to the HIV-1-LTR–RATS in a dose-dependent manner. To assess Ets-2 binding to RATS in vivo, Jurkat cells were transfected with 2× RATS-CAT and stained for the Ets-2 protein and the RATS sequence by combining immunofluorescence and fluorescence in situ hybridization techniques. In unstimulated cells, Ets-2 bound to RATS, whereas no binding was observed in stimulated cells. To test for RATS specificity, the same experiments were performed with 2× mutantRATS-CAT, and no binding of Ets-2 was observed. The results were corroborated by chromatin immunoprecipitation assays performed with the same cells. Our results show that Ets-2 is a transcriptional repressor of HIV-1. Repression of HIV-LTR-RATS mediated by Ets-2 may account for the low-level transcription and replication of HIV-1 in naive Th-cells, and contribute to the viral latency and maintenance of viral reservoirs in patients, despite long-term therapy.
Collapse
Affiliation(s)
- Ioannis Panagoulias
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Fotios Karagiannis
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Ioanna Aggeletopoulou
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Tassos Georgakopoulos
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| | - Christos P Argyropoulos
- Division of Nephrology, Department of Internal Medicine, Medical School, University of New Mexico, Albuquerque, NM, United States
| | - Karolina Akinosoglou
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Charalambos Gogos
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Athanasios Skoutelis
- Department of Internal Medicine and Infectious Diseases Unit, Evangelismos General Hospital, Athens, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras, Greece
| |
Collapse
|
13
|
Guo Y, MacIsaac KD, Chen Y, Miller RJ, Jain R, Joyce-Shaikh B, Ferguson H, Wang IM, Cristescu R, Mudgett J, Engstrom L, Piers KJ, Baltus GA, Barr K, Zhang H, Mehmet H, Hegde LG, Hu X, Carter LL, Aicher TD, Glick G, Zaller D, Hawwari A, Correll CC, Jones DC, Cua DJ. Inhibition of RORγT Skews TCRα Gene Rearrangement and Limits T Cell Repertoire Diversity. Cell Rep 2017; 17:3206-3218. [PMID: 28009290 DOI: 10.1016/j.celrep.2016.11.073] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/20/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
Recent studies have elucidated the molecular mechanism of RORγT transcriptional regulation of Th17 differentiation and function. RORγT was initially identified as a transcription factor required for thymopoiesis by maintaining survival of CD4+CD8+ (DP) thymocytes. While RORγ antagonists are currently being developed to treat autoimmunity, it remains unclear how RORγT inhibition may impact thymocyte development. In this study, we show that in addition to regulating DP thymocytes survival, RORγT also controls genes that regulate thymocyte migration, proliferation, and T cell receptor (TCR)α selection. Strikingly, pharmacological inhibition of RORγ skews TCRα gene rearrangement, limits T cell repertoire diversity, and inhibits development of autoimmune encephalomyelitis. Thus, targeting RORγT not only inhibits Th17 cell development and function but also fundamentally alters thymic-emigrant recognition of self and foreign antigens. The analysis of RORγ inhibitors has allowed us to gain a broader perspective of the diverse function of RORγT and its impact on T cell biology.
Collapse
Affiliation(s)
- Yanxia Guo
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - Kenzie D MacIsaac
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Yi Chen
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - Richard J Miller
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Renu Jain
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | | | - Heidi Ferguson
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - I-Ming Wang
- Merck Research Laboratories, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Razvan Cristescu
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - John Mudgett
- Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Laura Engstrom
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kyle J Piers
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Gretchen A Baltus
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kenneth Barr
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Hongjun Zhang
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Huseyin Mehmet
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | - Xiao Hu
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Laura L Carter
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | | | - Gary Glick
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Dennis Zaller
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Abbas Hawwari
- King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City Hospital, Ministry of National Guard Health Affairs, Mail Code 520, P.O. Box 6664, Al Hasa 31982, Kingdom of Saudi Arabia
| | - Craig C Correll
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Dallas C Jones
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Daniel J Cua
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA.
| |
Collapse
|
14
|
Pajcini KV, Xu L, Shao L, Petrovic J, Palasiewicz K, Ohtani Y, Bailis W, Lee C, Wertheim GB, Mani R, Muthusamy N, Li Y, Meijerink JPP, Blacklow SC, Faryabi RB, Cherry S, Pear WS. MAFB enhances oncogenic Notch signaling in T cell acute lymphoblastic leukemia. Sci Signal 2017; 10:10/505/eaam6846. [PMID: 29138297 DOI: 10.1126/scisignal.aam6846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Activating mutations in the gene encoding the cell-cell contact signaling protein Notch1 are common in human T cell acute lymphoblastic leukemias (T-ALLs). However, expressing Notch1 mutant alleles in mice fails to efficiently induce the development of leukemia. We performed a gain-of-function screen to identify proteins that enhanced signaling by leukemia-associated Notch1 mutants. The transcription factors MAFB and ETS2 emerged as candidates that individually enhanced Notch1 signaling, and when coexpressed, they synergistically increased signaling to an extent similar to that induced by core components of the Notch transcriptional complex. In mouse models of T-ALL, MAFB enhanced leukemogenesis by the naturally occurring Notch1 mutants, decreased disease latency, and increased disease penetrance. Decreasing MAFB abundance in mouse and human T-ALL cells reduced the expression of Notch1 target genes, including MYC and HES1, and sustained MAFB knockdown impaired T-ALL growth in a competitive setting. MAFB bound to ETS2 and interacted with the acetyltransferases PCAF and P300, highlighting its importance in recruiting coactivators that enhance Notch1 signaling. Together, these data identify a mechanism for enhancing the oncogenic potential of weak Notch1 mutants in leukemia models, and they reveal the MAFB-ETS2 transcriptional axis as a potential therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Kostandin V Pajcini
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA. .,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lanwei Xu
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lijian Shao
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jelena Petrovic
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karol Palasiewicz
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yumi Ohtani
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Will Bailis
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Curtis Lee
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajeswaran Mani
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Natarajan Muthusamy
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yunlei Li
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert B Faryabi
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Kotawong K, Thitapakorn V, Roytrakul S, Phaonakrop N, Viyanant V, Na-Bangchang K. Plasma Peptidome as a Source of Biomarkers for Diagnosis of Cholangiocarcinoma. Asian Pac J Cancer Prev 2017; 17:1163-8. [PMID: 27039742 DOI: 10.7314/apjcp.2016.17.3.1163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cholangiocarcinoma (CCA) is the bile duct cancer which constitutes one of the important public health problems in Thailand with high mortality rate, especially in the Opisthorchis viverrini (a parasite risk factor for CCA) endemic area of the northeastern region of the country. This study aimed to identify potential biomarkers from the plasma peptidome by CCA patients. Peptides were isolated using 10 kDa cut-off filter column and the flow-through was then used as a peptidome for LC-MS/MS analysis. A total of 209 peptides were obtained. Among these, 15 peptides were concerned with signaling pathways and 12 related to metabolic, regulatory, and biosynthesis of secondary metabolite pathways. Five exclusive peptides were identified as potential biomarkers, i.e. ETS domain-containing transcription factor ERF (P50548), KIAA0220 (Q92617), phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit beta isoform isoform 1 (P42338), LP2209 (Q6XYC0), and casein kinase II subunit alpha (P19784). Three of these biomarkers are signaling related molecules. A combination of these biomarkers for CCA diagnosis is proposed.
Collapse
Affiliation(s)
- Kanawut Kotawong
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, Thailand E-mail :
| | | | | | | | | | | |
Collapse
|
16
|
Panagoulias I, Georgakopoulos T, Aggeletopoulou I, Agelopoulos M, Thanos D, Mouzaki A. Transcription Factor Ets-2 Acts as a Preinduction Repressor of Interleukin-2 (IL-2) Transcription in Naive T Helper Lymphocytes. J Biol Chem 2016; 291:26707-26721. [PMID: 27815505 DOI: 10.1074/jbc.m116.762179] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/01/2016] [Indexed: 11/06/2022] Open
Abstract
IL-2 is the first cytokine produced when naive T helper (Th) cells are activated and differentiate into dividing pre-Th0 proliferating precursors. IL-2 expression is blocked in naive, but not activated or memory, Th cells by the transcription factor Ets-2 that binds to the antigen receptor response element (ARRE)-2 of the proximal IL-2 promoter. Ets-2 acts as an independent preinduction repressor in naive Th cells and does not interact physically with the transcription factor NFAT (nuclear factor of activated T-cells) that binds to the ARRE-2 in activated Th cells. In naive Th cells, Ets-2 mRNA expression, Ets-2 protein levels, and Ets-2 binding to ARRE-2 decrease upon cell activation followed by the concomitant expression of IL-2. Cyclosporine A stabilizes Ets-2 mRNA and protein when the cells are activated. Ets-2 silences directly constitutive or induced IL-2 expression through the ARRE-2. Conversely, Ets-2 silencing allows for constitutive IL-2 expression in unstimulated cells. Ets-2 binding to ARRE-2 in chromatin is stronger in naive compared with activated or memory Th cells; in the latter, Ets-2 participates in a change of the IL-2 promoter architecture, possibly to facilitate a quick response when the cells re-encounter antigen. We propose that Ets-2 expression and protein binding to the ARRE-2 of the IL-2 promoter are part of a strictly regulated process that results in a physiological transition of naive Th cells to Th0 cells upon antigenic stimulation. Malfunction of such a repression mechanism at the molecular level could lead to a disturbance of later events in Th cell plasticity, leading to autoimmune diseases or other pathological conditions.
Collapse
Affiliation(s)
- Ioannis Panagoulias
- From the Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras GR-26500, Greece and
| | - Tassos Georgakopoulos
- From the Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras GR-26500, Greece and
| | - Ioanna Aggeletopoulou
- From the Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras GR-26500, Greece and
| | - Marios Agelopoulos
- the Institute of Molecular Biology, Genetics and Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens GR-11527, Greece
| | - Dimitris Thanos
- the Institute of Molecular Biology, Genetics and Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens GR-11527, Greece
| | - Athanasia Mouzaki
- From the Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras GR-26500, Greece and
| |
Collapse
|
17
|
Liu X, Yan D, Li Y, Sha X, Wu K, Zhao J, Yang C, Zhang C, Shi J, Wu X. Erythroblast transformation-specific 2 correlates with vascular smooth muscle cell apoptosis in rat heterotopic heart transplantation model. J Thorac Dis 2016; 8:2027-37. [PMID: 27621856 DOI: 10.21037/jtd.2016.07.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) decreases the long-term survival of heart transplantation recipients. Vascular smooth muscle cell (VSMC) apoptosis is an important pathological feature of CAV. Erythroblast transformation-specific 2 (Ets-2), as a transcription factor, participates in cell apoptosis and plays an important role in organ transplantation. METHODS Hearts from Wistar-Furth (WF:RT1u) rats were heterotopically transplanted into Lewis (Lew:RT1(l)) rats without immunosuppression. Additional syngeneic heterotopic cardiac transplantations were performed in Lewis rats. HE staining was used to identify CAV. Ets-2 expression was examined by western blot. Ets-2 tissue location was examined by immunohistochemical assay and double immunostaining. Cleaved caspase 3 expression was detected by western blot. Co-localization of Ets-2 and cleaved caspase 3 was detected by double immunostaining. Ets-2, p53, cleaved caspase 3 and Bcl-xl expression in rat VSMC line A7R5 was examined after Ets-2 siRNA transfection. TUNEL assay was applied to detect A7R5 apoptosis with or without ETS-2 siRNA transfection. Immunoprecipitation was performed to explore the interaction between Ets-2 and p53. RESULTS Ets-2 expression decreased in the allograft group but had no obvious change in the isograft group. Meanwhile, the phenomenon of CAV was observed in the allograft group and there is neointima formation in the isograft group which is not obvious compared with allograft group. Additionally, Ets-2 expression was opposite to VSMC apoptosis in the allograft group. In vitro, Ets-2 siRNA transfection in A7R5cells resulted in enhanced cell apoptosis. Finally, Ets-2 interacted with p53. CONCLUSIONS Ets-2 might inhibit VSMC apoptosis via p53 pathway. The results further elucidate the molecular mechanism of VSMC apoptosis after heart transplantation during CAV and provide theoretical basis for seeking new specific drug targets for CAV prevention and treatment.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong 226001, China;; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China
| | - Daliang Yan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yangcheng Li
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Cancer Hospital of Nantong University, Nantong 226361, China
| | - Xilin Sha
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Rugao People's Hospital, Rugao 226500, China
| | - Kunpeng Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jianhua Zhao
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Chen Yang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Chao Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Vasculocardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jiahai Shi
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Xiang Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, China;; Department of Vasculocardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| |
Collapse
|
18
|
Lakshmi R, Jayavardhanan KK, Aravindakshan TV. Characterization of promoter sequence of toll-like receptor genes in Vechur cattle. Vet World 2016; 9:626-32. [PMID: 27397987 PMCID: PMC4937055 DOI: 10.14202/vetworld.2016.626-632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/18/2016] [Indexed: 12/02/2022] Open
Abstract
Aim: To analyze the promoter sequence of toll-like receptor (TLR) genes in Vechur cattle, an indigenous breed of Kerala with the sequence of Bos taurus and access the differences that could be attributed to innate immune responses against bovine mastitis. Materials and Methods: Blood samples were collected from Jugular vein of Vechur cattle, maintained at Vechur cattle conservation center of Kerala Veterinary and Animal Sciences University, using an acid-citrate-dextrose anticoagulant. The genomic DNA was extracted, and polymerase chain reaction was carried out to amplify the promoter region of TLRs. The amplified product of TLR2, 4, and 9 promoter regions was sequenced by Sanger enzymatic DNA sequencing technique. Results: The sequence of promoter region of TLR2 of Vechur cattle with the B. taurus sequence present in GenBank showed 98% similarity and revealed variants for four sequence motifs. The sequence of the promoter region of TLR4 of Vechur cattle revealed 99% similarity with that of B. taurus sequence but not reveals significant variant in motifregions. However, two heterozygous loci were observed from the chromatogram. Promoter sequence of TLR9 gene also showed 99% similarity to B. taurus sequence and revealed variants for four sequence motifs. Conclusion: The results of this study indicate that significant variation in the promoter of TLR2 and 9 genes in Vechur cattle breed and may potentially link the influence the innate immunity response against mastitis diseases.
Collapse
Affiliation(s)
- R Lakshmi
- Department of Veterinary Biochemistry, College of Veterinary and Animal Sciences, Thrissur, Kerala, India
| | - K K Jayavardhanan
- Department of Veterinary Biochemistry, College of Veterinary and Animal Sciences, Thrissur, Kerala, India
| | - T V Aravindakshan
- Centre for Advanced Studies in Animal Genetics and Breeding, College of Veterinary and Animal Sciences, Thrissur, Kerala, India
| |
Collapse
|
19
|
Murase R, Sato H, Yamamoto K, Ushida A, Nishito Y, Ikeda K, Kobayashi T, Yamamoto T, Taketomi Y, Murakami M. Group X Secreted Phospholipase A2 Releases ω3 Polyunsaturated Fatty Acids, Suppresses Colitis, and Promotes Sperm Fertility. J Biol Chem 2016; 291:6895-911. [PMID: 26828067 DOI: 10.1074/jbc.m116.715672] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Indexed: 12/31/2022] Open
Abstract
Within the secreted phospholipase A2(sPLA2) family, group X sPLA2(sPLA2-X) has the highest capacity to hydrolyze cellular membranes and has long been thought to promote inflammation by releasing arachidonic acid, a precursor of pro-inflammatory eicosanoids. Unexpectedly, we found that transgenic mice globally overexpressing human sPLA2-X (PLA2G10-Tg) displayed striking immunosuppressive and lean phenotypes with lymphopenia and increased M2-like macrophages, accompanied by marked elevation of free ω3 polyunsaturated fatty acids (PUFAs) and their metabolites. Studies usingPla2g10-deficient mice revealed that endogenous sPLA2-X, which is highly expressed in the colon epithelium and spermatozoa, mobilized ω3 PUFAs or their metabolites to protect against dextran sulfate-induced colitis and to promote fertilization, respectively. In colitis, sPLA2-X deficiency increased colorectal expression of Th17 cytokines, and ω3 PUFAs attenuated their production by lamina propria cells partly through the fatty acid receptor GPR120. In comparison, cytosolic phospholipase A2(cPLA2α) protects from colitis by mobilizing ω6 arachidonic acid metabolites, including prostaglandin E2 Thus, our results underscore a previously unrecognized role of sPLA2-X as an ω3 PUFA mobilizerin vivo, segregated mobilization of ω3 and ω6 PUFA metabolites by sPLA2-X and cPLA2α, respectively, in protection against colitis, and the novel role of a particular sPLA2-X-driven PUFA in fertilization.
Collapse
Affiliation(s)
- Remi Murase
- From the Lipid Metabolism Project and School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | | | | | - Ayako Ushida
- From the Lipid Metabolism Project and Department of Biology, Faculty of Science, Ochanomizu University, Tokyo 112-8610, Japan
| | - Yasumasa Nishito
- Core Technology and Research Center, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan, and
| | - Tetsuyuki Kobayashi
- Department of Biology, Faculty of Science, Ochanomizu University, Tokyo 112-8610, Japan
| | | | | | - Makoto Murakami
- From the Lipid Metabolism Project and AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
20
|
Fisher IB, Ostrowski M, Muthusamy N. Role for Ets-2(Thr-72) transcription factor in stage-specific thymocyte development and survival. J Biol Chem 2011; 287:5199-210. [PMID: 22128184 DOI: 10.1074/jbc.m111.242602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Interference of Ras signaling deregulates thymocyte development in mouse models. However, the role of Ets-2, a transcription factor that is phosphorylated on a critical threonine residue (Thr-72) by the Ras/MAPK pathway in thymocyte development, has not been defined. Transgenic mice overexpressing a phosphomutant Ets-2 (T72A) in the thymus displayed reduced thymus size associated with a 60-80% reduction in thymocyte populations. The transgenic mice exhibited a 20-fold increase in a c-Kit(+) CD4(+) CD8(+) CD3(-) population and a 5-fold increase in a unique CD5(low) population associated with a partial developmental block at the DN2-DN3 stage of thymocytes. Transgenic thymocytes exhibited increased apoptosis, and overexpression of Bcl-2 rescued the hypocellularity and associated thymocyte developmental block in double transgenic mice. The observed defects in these mice are not dependent on Ets-1 expression. These studies implicate for the first time a stage-specific Ets-1-independent regulatory role for Ets-2 in early thymocyte development and survival.
Collapse
Affiliation(s)
- Ian B Fisher
- Molecular Cellular Developmental Biology Program, Division of Hematology, Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
21
|
Shi Z, Cai Z, Wen S, Chen C, Gendron C, Sanchez A, Patterson K, Fu S, Yang J, Wildman D, Finnell RH, Zhang D. Transcriptional regulation of the novel Toll-like receptor Tlr13. J Biol Chem 2009; 284:20540-7. [PMID: 19487701 DOI: 10.1074/jbc.m109.022541] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Little has been known about Tlr13 (Toll-like receptor 13), a novel member of the Toll-like receptor family. To elucidate the molecular basis of murine Tlr13 gene expression, the activity of the Tlr13 gene promoter was characterized. Reporter gene analysis and electrophoretic mobility shift assays demonstrated that Tlr13 gene transcription was regulated through three cis-acting elements that interacted with the Ets2, Sp1, and PU.1 transcription factors. Furthermore, our work suggests that these transcription factors may cooperate, culminating in maximal transcription of the Tlr13 gene. In contrast, NF-kappaB appeared to act as an inhibitor of Tlr13 transcription. Overexpression of Ets2 caused a strong increase in the transcriptional activity of the Tlr13 promoter; however, overexpression of NF-kappaB p65 dramatically inhibited it. Additionally, interferon-beta is capable of acting Tlr13 transcription, but the activated signaling of lipopolysaccharide/TLR4 and peptidoglycan/TLR2 strongly inhibited the Tlr13 gene promoter. Thus, these findings reveal the mechanism of Tlr13 gene regulation, thereby providing insight into the function of Tlr13 in the immune response to pathogen.
Collapse
Affiliation(s)
- Zhongcheng Shi
- Center for Infectious and Inflammatory Disease, Institute of Bioscience and Technology, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kuang X, Shen J, Wong PK, Yan M. Deregulation of mTOR signaling is involved in thymic lymphoma development in Atm−/− mice. Biochem Biophys Res Commun 2009; 383:368-72. [DOI: 10.1016/j.bbrc.2009.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 04/03/2009] [Indexed: 12/18/2022]
|
23
|
Okamura K, Yamashita S, Ando H, Horibata Y, Aoyama C, Takagishi K, Izumi T, Vance DE, Sugimoto H. Identification of nuclear localization and nuclear export signals in Ets2, and the transcriptional regulation of Ets2 and CTP:phosphocholine cytidylyltransferase α in tetradecanoyl-13-acetate or macrophage-colony stimulating factor stimulated RAW264 cells. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:173-82. [DOI: 10.1016/j.bbalip.2008.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 12/16/2008] [Accepted: 12/18/2008] [Indexed: 11/24/2022]
|
24
|
David-Fung ES, Butler R, Buzi G, Yui MA, Diamond RA, Anderson MK, Rowen L, Rothenberg EV. Transcription factor expression dynamics of early T-lymphocyte specification and commitment. Dev Biol 2008; 325:444-67. [PMID: 19013443 DOI: 10.1016/j.ydbio.2008.10.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 10/17/2008] [Indexed: 12/15/2022]
Abstract
Mammalian T lymphocytes are a prototype for development from adult pluripotent stem cells. While T-cell specification is driven by Notch signaling, T-lineage commitment is only finalized after prolonged Notch activation. However, no T-lineage specific regulatory factor has been reported that mediates commitment. We used a gene-discovery approach to identify additional candidate T-lineage transcription factors and characterized expression of >100 regulatory genes in early T-cell precursors using realtime RT-PCR. These regulatory genes were also monitored in multilineage precursors as they entered T-cell or non-T-cell pathways in vitro; in non-T cells ex vivo; and in later T-cell developmental stages after lineage commitment. At least three major expression patterns were observed. Transcription factors in the largest group are expressed at relatively stable levels throughout T-lineage specification as a legacy from prethymic precursors, with some continuing while others are downregulated after commitment. Another group is highly expressed in the earliest stages only, and is downregulated before or during commitment. Genes in a third group undergo upregulation at one of three distinct transitions, suggesting a positive regulatory cascade. However, the transcription factors induced during commitment are not T-lineage specific. Different members of the same transcription factor family can follow opposite trajectories during specification and commitment, while factors co-expressed early can be expressed in divergent patterns in later T-cell development. Some factors reveal new regulatory distinctions between alphabeta and gammadelta T-lineage differentiation. These results show that T-cell identity has an essentially complex regulatory basis and provide a detailed framework for regulatory network modeling of T-cell specification.
Collapse
|
25
|
Ge Y, LaFiura KM, Dombkowski AA, Chen Q, Payton SG, Buck SA, Salagrama S, Diakiw AE, Matherly LH, Taub JW. The role of the proto-oncogene ETS2 in acute megakaryocytic leukemia biology and therapy. Leukemia 2007; 22:521-9. [PMID: 18094719 DOI: 10.1038/sj.leu.2405066] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Acute myeloid leukemia (AML) in Down syndrome (DS) children has several unique features including a predominance of the acute megakaryocytic leukemia (AMkL) phenotype, higher event-free survivals compared to non-DS children using cytosine arabinoside (ara-C)/anthracycline-based protocols and a uniform presence of somatic mutations in the X-linked transcription factor gene, GATA1. Several chromosome 21-localized transcription factor oncogenes including ETS2 may contribute to the unique features of DS AMkL. ETS2 transcripts measured by real-time RT-PCR were 1.8- and 4.1-fold, respectively, higher in DS and non-DS megakaryoblasts than those in non-DS myeloblasts. In a doxycycline-inducible erythroleukemia cell line, K562pTet-on/ETS2, induction of ETS2 resulted in an erythroid to megakaryocytic phenotypic switch independent of GATA1 levels. Microarray analysis of doxycycline-induced and doxycycline-uninduced cells revealed an upregulation by ETS2 of cytokines (for example, interleukin 1 and CSF2) and transcription factors (for example, TAL1), which are key regulators of megakaryocytic differentiation. In the K562pTet-on/ETS2 cells, ETS2 induction conferred differences in sensitivities to ara-C and daunorubicin, depending on GATA1 levels. These results suggest that ETS2 expression is linked to the biology of AMkL in both DS and non-DS children, and that ETS2 acts by regulating expression of hematopoietic lineage and transcription factor genes involved in erythropoiesis and megakaryopoiesis, and in chemotherapy sensitivities.
Collapse
Affiliation(s)
- Y Ge
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
David-Fung ES, Yui MA, Morales M, Wang H, Taghon T, Diamond RA, Rothenberg EV. Progression of regulatory gene expression states in fetal and adult pro-T-cell development. Immunol Rev 2006; 209:212-36. [PMID: 16448545 PMCID: PMC4157939 DOI: 10.1111/j.0105-2896.2006.00355.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Precursors entering the T-cell developmental pathway traverse a progression of states characterized by distinctive patterns of gene expression. Of particular interest are regulatory genes, which ultimately control the dwell time of cells in each state and establish the mechanisms that propel them forward to subsequent states. Under particular genetic and developmental circumstances, the transitions between these states occur with different timing, and environmental feedbacks may shift the steady-state accumulations of cells in each state. The fetal transit through pro-T-cell stages is faster than in the adult and subject to somewhat different genetic requirements. To explore causes of such variation, this review presents previously unpublished data on differentiation gene activation in pro-T cells of pre-T-cell receptor-deficient mutant mice and a quantitative comparison of the profiles of transcription factor gene expression in pro-T-cell subsets of fetal and adult wildtype mice. Against a background of consistent gene expression, several regulatory genes show marked differences between fetal and adult expression profiles, including those encoding two basic helix-loop-helix antagonist Id factors, the Ets family factor SpiB and the Notch target gene Deltex1. The results also reveal global differences in regulatory alterations triggered by the first T-cell receptor-dependent selection events in fetal and adult thymopoiesis.
Collapse
|
27
|
Abstract
Transcriptional regulation of T-cell development involves successive interactions between complexes of transcriptional regulators and their binding sites within the regulatory regions of each gene. The regulatory modules that control expression of T-lineage genes frequently include binding sites for a core set of regulators that set the T-cell-specific background for signal-dependent control, including GATA-3, Notch/CSL, c-myb, TCF-1, Ikaros, HEB/E2A, Ets, and Runx factors. Additional regulators in early thymocytes include PU.1, Id-2, SCL, Spi-B, Erg, Gfi-1, and Gli. Many of these factors are involved in simultaneous regulation of non-T-lineage genes, T-lineage genes, and genes involved in cell cycle control, apoptosis, or survival. Potential and known interactions between early thymic transcription factors such as GATA-3, SCL, PU.1, Erg, and Spi-B are explored. Regulatory modules involved in the expression of several critical T-lineage genes are described, and models are presented for shifting occupancy of the DNA-binding sites in the regulatory modules of pre-Talpha, T-cell receptor beta (TCRbeta), recombinase activating genes 1 and 2 (Rag-1/2), and CD4 during T-cell development. Finally, evidence is presented that c-kit, Erg, Hes-1, and HEBAlt are expressed differently in Rag-2(-/-) thymocytes versus normal early thymocytes, which provide insight into potential regulatory interactions that occur during normal T-cell development.
Collapse
Affiliation(s)
- Michele K Anderson
- Sunnybrook and Women's College Health Sciences Center, Division of Molecular and Cell Biology, University of Toronto, Department of Immunology, Toronto, ON, Canada.
| |
Collapse
|
28
|
Zhang Q, Badell IR, Schwarz EM, Boulukos KE, Yao Z, Boyce BF, Xing L. Tumor necrosis factor prevents alendronate-induced osteoclast apoptosis in vivo by stimulating Bcl-xL expression through Ets-2. ACTA ACUST UNITED AC 2005; 52:2708-18. [PMID: 16142752 DOI: 10.1002/art.21236] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To investigate why bisphosphonates are less effective at preventing focal bone loss in rheumatoid arthritis (RA) patients than in those with generalized osteoporosis, and the mechanisms involved. METHODS The response of osteoclasts to alendronate (ALN) in tumor necrosis factor-transgenic (TNF-Tg) mice that develop erosive arthritis and in wild-type littermates was studied. TNF-Tg and wild-type mice were given ALN, and the osteoclast numbers in the inflamed joints and in the long bones were compared. The expression levels of Bcl-xL in the osteoclasts of TNF-Tg and wild-type mice were examined by immunostaining. The effect of overexpression of Bcl-xL and Ets-2 proteins on ALN-induced osteoclast apoptosis was determined using an in vitro osteoclast survival assay and retrovirus transfer approach. RESULTS ALN reduced osteoclast numbers in the metaphyses by 97%, but by only 46% in the adjacent inflamed joints. Bcl-xL expression was markedly higher in osteoclasts in the joints than in those in the metaphyses of TNF-Tg mice. Bcl-xL or Ets-2 overexpression protected osteoclasts from ALN-induced apoptosis, and TNF stimulated Bcl-xL and Ets-2 expression in osteoclasts. Overexpression of Ets-2 increased Bcl-xL messenger RNA in osteoclasts, while a dominant-negative form of the Ets-2 blocked the protective effect of Bcl-xL or TNF on ALN-induced apoptosis. CONCLUSION The reduced efficacy of bisphosphonates to stop bone erosion in the inflamed joints of RA patients may result from local high levels of TNF up-regulating Ets-2 expression in osteoclasts, which in turn stimulates Bcl-xL expression in them and reduces their susceptibility to bisphosphonate-induced apoptosis.
Collapse
Affiliation(s)
- Q Zhang
- University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Kawagoe H, Grosveld GC. MN1-TEL myeloid oncoprotein expressed in multipotent progenitors perturbs both myeloid and lymphoid growth and causes T-lymphoid tumors in mice. Blood 2005; 106:4278-86. [PMID: 16081688 PMCID: PMC1895241 DOI: 10.1182/blood-2005-04-1674] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MN1-TEL (meningioma 1-translocation-ETS-leukemia) fusion oncoprotein is the product of the t(12;22)(p13;q11) in human myeloid leukemia consisting of N-terminal MN1 sequences, a transcriptional coactivator, fused to C-terminal TEL sequences, an E26-transformation-specific (ETS) transcription factor. To analyze the role of MN1-TEL in leukemogenesis, we created a site-directed transgenic (knock-in) mouse model carrying a conditional MN1-TEL transgene under the control of the Aml1 regulatory sequences. After induction, MN1-TEL expression was detected in both myeloid and lymphoid cells. Activation of MN1-TEL expression enhanced the repopulation ability of myeloid progenitors in vitro as well as partially inhibited their differentiation in vivo. MN1-TEL also promoted the proliferation of thymocytes while it blocked their differentiation from CD4-/CD8- to CD4+/CD8+ in vivo. After long latency, 30% of the MN1-TEL-positive mice developed T-lymphoid tumors. This process was accelerated by N-ethyl-N-nitrosourea-induced mutations. MN1-TEL-positive T-lymphoid tumors showed elevated expression of the Notch-1, Hes-1, c-Myc, and Lmo-2 genes while their Ink4a/pRB and Arf/p53 pathways were impaired, suggesting that these alterations cooperatively transform T progenitors. We conclude that MN1-TEL exerts its nonlineage-specific leukemogenic effects by promoting the growth of primitive progenitors and blocking their differentiation, but cooperative mutations are necessary to fully induce leukemic transformation.
Collapse
Affiliation(s)
- Hiroyuki Kawagoe
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA
| | | |
Collapse
|
30
|
Takeshita F, Suzuki K, Sasaki S, Ishii N, Klinman DM, Ishii KJ. Transcriptional Regulation of the Human TLR9 Gene. THE JOURNAL OF IMMUNOLOGY 2004; 173:2552-61. [PMID: 15294971 DOI: 10.4049/jimmunol.173.4.2552] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To clarify the molecular basis of human TLR9 (hTLR9) gene expression, the activity of the hTLR9 gene promoter was characterized using the human myeloma cell line RPMI 8226. Reporter gene analysis and EMSA demonstrated that hTLR9 gene transcription was regulated via four cis-acting elements, cAMP response element, 5'-PU box, 3'-PU box, and a C/EBP site, that interacted with the CREB1, Ets2, Elf1, Elk1, and C/EBPalpha transcription factors. Other members of the C/EBP family, such as C/EBPbeta, C/EBPdelta, and C/EBPepsilon, were also important for TLR9 gene transcription. CpG DNA-mediated suppression of TLR9 gene transcription led to decreased binding of the trans-acting factors to their corresponding cis-acting elements. It appeared that suppression was mediated via c-Jun and NF-kappaB p65 and that cooperation among CREB1, Ets2, Elf1, Elk1, and C/EBPalpha culminated in maximal transcription of the TLR9 gene. These findings will help to elucidate the mechanism of TLR9 gene regulation and to provide insight into the process by which TLR9 evolved in the mammalian immune system.
Collapse
MESH Headings
- Animals
- Base Sequence
- Blotting, Western
- CCAAT-Enhancer-Binding Proteins/immunology
- CCAAT-Enhancer-Binding Proteins/metabolism
- Cell Line, Tumor
- Cloning, Molecular
- CpG Islands/genetics
- CpG Islands/immunology
- Cyclic AMP Response Element-Binding Protein/immunology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Gene Expression Regulation/immunology
- Genes, Reporter/genetics
- Genes, Reporter/immunology
- Genes, jun/immunology
- Humans
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- NF-kappa B/immunology
- NF-kappa B/metabolism
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/immunology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Nucleic Acid
- Toll-Like Receptor 9
- Toll-Like Receptors
- Transcription Factors/immunology
- Transcription Factors/metabolism
- Transcription, Genetic/genetics
- Transcriptional Activation/immunology
- Transfection
Collapse
Affiliation(s)
- Fumihiko Takeshita
- Section of Retroviral Immunology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Leider N, Melamed D. Differential c-Myc responsiveness to B cell receptor ligation in B cell-negative selection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2446-52. [PMID: 12928392 DOI: 10.4049/jimmunol.171.5.2446] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Responsiveness of c-Myc oncogene to B cell receptor ligation has been implicated in the induction of apoptosis in transformed and normal immature B cells. These studies provided compelling evidence to link the c-Myc oncogene with the process of negative selection in B-lymphocytes. However, in addition to apoptosis, B cell-negative selection has been shown to occur by secondary Ig gene rearrangements, a mechanism called receptor editing. In this study, we assessed whether differential c-Myc responsiveness to B cell receptor (BCR) ligation is associated with the mechanism of negative selection in immature B cells. Using an in vitro bone marrow culture system and an Ig-transgenic mouse model (3-83) we show here that c-Myc is expressed at low levels throughout B cell development and that c-Myc responsiveness to BCR ligation is developmentally regulated and increased with maturation. Furthermore, we found that the competence to mount c-Myc responsiveness upon BCR ligation is important for the induction of apoptosis and had no effect on the process of receptor editing. Therefore, this study suggests an important role of c-Myc in promoting and/or maintaining B cell development and that compartmentalization of B cell tolerance may also be developmentally regulated by differential c-Myc responsiveness.
Collapse
MESH Headings
- Animals
- Antigens, CD19/genetics
- Apoptosis/genetics
- Apoptosis/immunology
- B-Lymphocyte Subsets/cytology
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line, Transformed
- Cells, Cultured
- DNA-Binding Proteins/biosynthesis
- Gene Expression Regulation/immunology
- Genes, myc/immunology
- Ligands
- Lymphopoiesis/genetics
- Lymphopoiesis/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Oligonucleotides, Antisense/pharmacology
- Proto-Oncogene Proteins c-myc/biosynthesis
- Proto-Oncogene Proteins c-myc/genetics
- RNA Editing/genetics
- RNA Editing/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Self Tolerance/genetics
Collapse
Affiliation(s)
- Nira Leider
- Department of Immunology, Technion Faculty of Medicine, Haifa 31096, Israel
| | | |
Collapse
|