1
|
Wang J, Wang X, Guo Y, Ye L, Li D, Hu A, Cai S, Yuan B, Jin S, Zhou Y, Li Q, Zheng L, Tong Q. Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancer cells and neutrophils inhibits aerobic glycolysis and cancer progression. Clin Transl Med 2021; 11:e588. [PMID: 34841706 PMCID: PMC8567044 DOI: 10.1002/ctm2.588] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND As a metabolic reprogramming feature, cancer cells derive most of their energy from aerobic glycolysis, while its regulatory mechanisms and therapeutic strategies continue to be illusive. METHODS Integrative analysis of publically available expression profile datasets was used to identify critical transcriptional regulators and their target glycolytic enzymes. The functions and acting mechanisms of transcriptional regulators in cancer cells were investigated by using in vitro and in vivo assays. The Kaplan-Meier curve and log-rank assay were used to conduct the survival study. RESULTS Salmonella pathogenicity island 1 (SPI1/PU.1), a haematopoietic transcription factor, was identified to facilitate glycolytic process, tumourigenesis, invasiveness, as well as metastasis of colon cancer cells, which was interplayed by tumour-associated neutrophils. Mechanistically, neutrophils delivered SPI1 mRNA via extracellular vesicles, resulting in enhanced SPI1 expression of cancer cells. Through physical interaction with SPI1-related protein (SPIB), SPI1 drove expression of glycolytic genes within cancer cells, which in turn induced polarization of neutrophils via glycolytic metabolite lactate. Depletion of neutrophils or SPIB-SPI1 interaction in cancer cells significantly inhibited glycolytic process, tumourigenesis and aggressiveness. Upregulation of SPI1 or SPIB was found to be associated with poor prognosis in patients suffering from colon cancer. CONCLUSIONS Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancerous cells and neutrophils suppresses aerobic glycolysis and progression of cancer.
Collapse
Affiliation(s)
- Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Xiaojing Wang
- Department of Geriatrics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yanhua Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Lin Ye
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Anpei Hu
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shuang Cai
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Boling Yuan
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shikai Jin
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yi Zhou
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| |
Collapse
|
2
|
Kaasinen E, Kuismin O, Rajamäki K, Ristolainen H, Aavikko M, Kondelin J, Saarinen S, Berta DG, Katainen R, Hirvonen EAM, Karhu A, Taira A, Tanskanen T, Alkodsi A, Taipale M, Morgunova E, Franssila K, Lehtonen R, Mäkinen M, Aittomäki K, Palotie A, Kurki MI, Pietiläinen O, Hilpert M, Saarentaus E, Niinimäki J, Junttila J, Kaikkonen K, Vahteristo P, Skoda RC, Seppänen MRJ, Eklund KK, Taipale J, Kilpivaara O, Aaltonen LA. Impact of constitutional TET2 haploinsufficiency on molecular and clinical phenotype in humans. Nat Commun 2019; 10:1252. [PMID: 30890702 PMCID: PMC6424975 DOI: 10.1038/s41467-019-09198-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Abstract
Clonal hematopoiesis driven by somatic heterozygous TET2 loss is linked to malignant degeneration via consequent aberrant DNA methylation, and possibly to cardiovascular disease via increased cytokine and chemokine expression as reported in mice. Here, we discover a germline TET2 mutation in a lymphoma family. We observe neither unusual predisposition to atherosclerosis nor abnormal pro-inflammatory cytokine or chemokine expression. The latter finding is confirmed in cells from three additional unrelated TET2 germline mutation carriers. The TET2 defect elevates blood DNA methylation levels, especially at active enhancers and cell-type specific regulatory regions with binding sequences of master transcription factors involved in hematopoiesis. The regions display reduced methylation relative to all open chromatin regions in four DNMT3A germline mutation carriers, potentially due to TET2-mediated oxidation. Our findings provide insight into the interplay between epigenetic modulators and transcription factor activity in hematological neoplasia, but do not confirm the putative role of TET2 in atherosclerosis.
Collapse
Affiliation(s)
- Eevi Kaasinen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, SE 171 77, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77, Stockholm, Sweden
| | - Outi Kuismin
- Department of Clinical Genetics, Oulu University Hospital, FI-90029, Oulu, Finland
- PEDEGO Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, FI-00014, Helsinki, Finland
| | - Kristiina Rajamäki
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
- Clinicum, University of Helsinki, FI-00014, Helsinki, Finland
| | - Heikki Ristolainen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Mervi Aavikko
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Johanna Kondelin
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Silva Saarinen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Davide G Berta
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Riku Katainen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Elina A M Hirvonen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Aurora Taira
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Tomas Tanskanen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Amjad Alkodsi
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Minna Taipale
- Department of Biosciences and Nutrition, Karolinska Institutet, SE 171 77, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77, Stockholm, Sweden
| | - Ekaterina Morgunova
- Department of Biosciences and Nutrition, Karolinska Institutet, SE 171 77, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77, Stockholm, Sweden
| | - Kaarle Franssila
- HUSLAB, Helsinki University Hospital, FI-00029, Helsinki, Finland
| | - Rainer Lehtonen
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Markus Mäkinen
- Cancer and Translational Medicine Research Unit, University of Oulu, FI-90014, Oulu, Finland
| | - Kristiina Aittomäki
- Department of Clinical Genetics, Helsinki University Hospital, FI-00029, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, FI-00014, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, 02114, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA
| | - Mitja I Kurki
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Olli Pietiläinen
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, 02142, MA, USA
| | - Morgane Hilpert
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, CH-4031, Switzerland
| | - Elmo Saarentaus
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, FI-00014, Helsinki, Finland
| | - Jaakko Niinimäki
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014, Oulu, Finland
| | - Juhani Junttila
- Research Unit of Internal Medicine, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland
| | - Kari Kaikkonen
- Research Unit of Internal Medicine, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, FI-90014, Oulu, Finland
| | - Pia Vahteristo
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
| | - Radek C Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, CH-4031, Switzerland
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, FI-00029, Helsinki, Finland
- Rare Diseases Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, FI-00029, Helsinki, Finland
| | - Kari K Eklund
- Clinicum, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Rheumatology, Helsinki University Hospital, FI-00029, Helsinki, Finland
- ORTON Orthopaedic Hospital, FI-00280, Helsinki, Finland
| | - Jussi Taipale
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, SE 171 77, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE 171 77, Stockholm, Sweden
| | - Outi Kilpivaara
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland.
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, University of Helsinki, FI-00014, Helsinki, Finland.
- Genome-Scale Biology, Research Programs Unit, University of Helsinki, FI-00014, Helsinki, Finland.
- Department of Biosciences and Nutrition, Karolinska Institutet, SE 171 77, Stockholm, Sweden.
| |
Collapse
|
3
|
|
4
|
Goswami R, Kaplan MH. Gcn5 is required for PU.1-dependent IL-9 induction in Th9 cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:3026-33. [PMID: 22904310 DOI: 10.4049/jimmunol.1201496] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Naive CD4+ T cells differentiate into various effector Th subsets depending on the Ags and cytokine microenvironment they encounter. IL-9-secreting Th9 cells are the most recent Th subset to be described. PU.1, one of the transcription factors required for the development of Th9 cells, binds to the Il9 gene. In this study, we show that PU.1 increases histone acetylation at the Il9 locus through direct interactions with histone acetyltransferases. In the absence of PU.1, there is decreased association of Gcn5 and p300/CBP associated factor and increased association of histone deacetylases at the Il9 locus in Th9 cells. Inhibition of histone deacetylase activity augments PU.1-dependent IL-9 production. PU.1 forms a complex with Gcn5, and inhibition of the expression of Gcn5 results in reduced IL-9 production. Moreover, the effects of Gcn5 on IL-9 production are specific as the production of IL-10 and IL-21, two additional cytokines produced by Th9 cells, is not altered after decreased Gcn5 expression. Together, these data define a PU.1-dependent mechanism for altered histone acetylation and expression of the Il9 locus in Th9 cells.
Collapse
Affiliation(s)
- Ritobrata Goswami
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
5
|
Escoubet-Lozach L, Benner C, Kaikkonen MU, Lozach J, Heinz S, Spann NJ, Crotti A, Stender J, Ghisletti S, Reichart D, Cheng CS, Luna R, Ludka C, Sasik R, Garcia-Bassets I, Hoffmann A, Subramaniam S, Hardiman G, Rosenfeld MG, Glass CK. Mechanisms establishing TLR4-responsive activation states of inflammatory response genes. PLoS Genet 2011; 7:e1002401. [PMID: 22174696 PMCID: PMC3234212 DOI: 10.1371/journal.pgen.1002401] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/13/2011] [Indexed: 01/22/2023] Open
Abstract
Precise control of the innate immune response is required for resistance to microbial infections and maintenance of normal tissue homeostasis. Because this response involves coordinate regulation of hundreds of genes, it provides a powerful biological system to elucidate the molecular strategies that underlie signal- and time-dependent transitions of gene expression. Comprehensive genome-wide analysis of the epigenetic and transcription status of the TLR4-induced transcriptional program in macrophages suggests that Toll-like receptor 4 (TLR4)-dependent activation of nearly all immediate/early- (I/E) and late-response genes results from a sequential process in which signal-independent factors initially establish basal levels of gene expression that are then amplified by signal-dependent transcription factors. Promoters of I/E genes are distinguished from those of late genes by encoding a distinct set of signal-dependent transcription factor elements, including TATA boxes, which lead to preferential binding of TBP and basal enrichment for RNA polymerase II immediately downstream of transcriptional start sites. Global nuclear run-on (GRO) sequencing and total RNA sequencing further indicates that TLR4 signaling markedly increases the overall rates of both transcriptional initiation and the efficiency of transcriptional elongation of nearly all I/E genes, while RNA splicing is largely unaffected. Collectively, these findings reveal broadly utilized mechanisms underlying temporally distinct patterns of TLR4-dependent gene activation required for homeostasis and effective immune responses. The innate immune response is a complex biological program that is configured to allow host cells to rapidly respond to infection and tissue injury. An essential feature of this response is the sequential activation of large numbers of genes that play roles in amplification of the initial inflammatory response, exert anti-microbial activities, and initiate acquired immunity. Here, we use a combination of genome-wide approaches to characterize the basal and activated states of promoters that drive the expression of genes that are turned on at immediate/early or late times in macrophages following their stimulation with a mimetic of bacterial infection. These studies identify genetically encoded features that establish basal levels of expression and distinct temporal profiles of signal-dependent gene activation required for effective immune responses. The general features of immediate/early and late genes defined by these studies are likely to be instructive for understanding how other high-magnitude, temporally orchestrated programs of gene expression are established.
Collapse
Affiliation(s)
- Laure Escoubet-Lozach
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Christopher Benner
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Minna U. Kaikkonen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- A. I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jean Lozach
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Sven Heinz
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Nathan J. Spann
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Andrea Crotti
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Josh Stender
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Serena Ghisletti
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Donna Reichart
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Christine S. Cheng
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Rosa Luna
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Colleen Ludka
- Biomedical Genomics Microarray Laboratory (BIOGEM), University of California San Diego, La Jolla, California, United States of America
| | - Roman Sasik
- Biomedical Genomics Microarray Laboratory (BIOGEM), University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Ivan Garcia-Bassets
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, California, United States of America
| | - Alexander Hoffmann
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Shankar Subramaniam
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Gary Hardiman
- Biomedical Genomics Microarray Laboratory (BIOGEM), University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Michael G. Rosenfeld
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, California, United States of America
| | - Christopher K. Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
6
|
The Transcription Factor PU.1 is a Critical Regulator of Cellular Communication in the Immune System. Arch Immunol Ther Exp (Warsz) 2011; 59:431-40. [DOI: 10.1007/s00005-011-0147-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/25/2011] [Indexed: 12/22/2022]
|
7
|
Choe KS, Ujhelly O, Wontakal SN, Skoultchi AI. PU.1 directly regulates cdk6 gene expression, linking the cell proliferation and differentiation programs in erythroid cells. J Biol Chem 2009; 285:3044-52. [PMID: 19955566 DOI: 10.1074/jbc.m109.077727] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell proliferation and differentiation are highly coordinated processes during normal development. Most leukemia cells are blocked from undergoing terminal differentiation and also exhibit uncontrolled proliferation. Dysregulated expression of transcription factor PU.1 is strongly associated with Friend virus-induced erythroleukemia. PU.1 inhibits erythroid differentiation by binding to and inhibiting GATA-1. PU.1 also may be involved in controlling proliferation of erythroid cells. We reported previously that the G(1) phase-specific cyclin-dependent kinase 6 (CDK6) also blocks erythroid differentiation. We now report that PU.1 directly stimulates transcription of the cdk6 gene in both normal erythroid progenitors and erythroleukemia cells, as well as in macrophages. We propose that PU.1 coordinates proliferation and differentiation in immature erythroid cells by inhibiting the GATA-1-mediated gene expression program and also by regulating expression of genes that control progression through the G(1) phase of the cell cycle, the period during which the decision to differentiate is made.
Collapse
Affiliation(s)
- Kevin S Choe
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | |
Collapse
|
8
|
The Promoter of the Immunoglobulin J Chain Gene Receives Its Authentic Enhancer Activity through the Abutting MEF2 and PU.1 Sites in a DNA-Looping Interaction. J Mol Biol 2009; 390:339-52. [DOI: 10.1016/j.jmb.2009.05.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 05/13/2009] [Accepted: 05/20/2009] [Indexed: 11/17/2022]
|
9
|
Gupta P, Gurudutta GU, Saluja D, Tripathi RP. PU.1 and partners: regulation of haematopoietic stem cell fate in normal and malignant haematopoiesis. J Cell Mol Med 2009; 13:4349-63. [PMID: 19382896 PMCID: PMC4515051 DOI: 10.1111/j.1582-4934.2009.00757.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
During normal haematopoiesis, cell development and differentiation programs are accomplished by switching ‘on’ and ‘off’ specific set of genes. Specificity of gene expression is primarily achieved by combinatorial control, i.e. through physical and functional interactions among several transcription factors that form sequence-specific multiprotein complexes on regulatory regions (gene promoters and enhancers). Such combinatorial gene switches permit flexibility of regulation and allow numerous developmental decisions to be taken with a limited number of regulators. The haematopoietic-specific Ets family transcription factor PU.1 regulates many lymphoid- and myeloid-specific gene promoters and enhancers by interacting with multiple proteins during haematopoietic development. Such protein–protein interactions regulate DNA binding, subcellular localization, target gene selection and transcriptional activity of PU.1 itself in response to diverse signals including cytokines, growth factors, antigen and cellular stresses. Specific domains of PU.1 interact with many protein motifs such as bHLH, bZipper, zinc fingers and paired domain for regulating its activity. This review focuses on important protein–protein interactions of PU.1 that play a crucial role in regulation of normal as well as malignant haematopoiesis. Precise delineation of PU.1 protein-partner interacting interface may provide an improved insight of the molecular mechanisms underlying haematopoietic stem cell fate regulation. Its interactions with some proteins could be targeted to modulate the aberrant signalling pathways for reversing the malignant phenotype and to control the generation of specific haematopoietic progeny for treatment of haematopoietic disorders.
Collapse
Affiliation(s)
- Pallavi Gupta
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, DRDO, Delhi, India
| | | | | | | |
Collapse
|
10
|
|
11
|
Schweitzer BL, Huang KJ, Kamath MB, Emelyanov AV, Birshtein BK, DeKoter RP. Spi-C has opposing effects to PU.1 on gene expression in progenitor B cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:2195-207. [PMID: 16887979 DOI: 10.4049/jimmunol.177.4.2195] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Ets transcription factor Spi-C, expressed in B cells and macrophages, is closely related to PU.1 and has the ability to recognize the same DNA consensus sequence. However, the function of Spi-C has yet to be determined. The purpose of this study is to further examine Spi-C activity in B cell development. First, using retroviral vectors to infect PU.1(-/-) fetal liver progenitors, Spi-C was found to be inefficient at inducing cytokine-dependent proliferation and differentiation of progenitor B (pro-B) cells or macrophages relative to PU.1 or Spi-B. Next, Spi-C was ectopically expressed in fetal liver-derived, IL-7-dependent pro-B cell lines. Wild-type (WT) pro-B cells ectopically expressing Spi-C (WT-Spi-C) have several phenotypic characteristics of pre-B cells such as increased CD25 and decreased c-Kit surface expression. In addition, WT-Spi-C pro-B cells express increased levels of IgH sterile transcripts and reduced levels of expression and transcription of the FcgammaRIIb gene. Gel-shift analysis suggests that Spi-C, ectopically expressed in pro-B cells, can bind PU.1 consensus sites in the IgH intronic enhancer and FcgammaRIIb promoter. Transient transfection analysis demonstrated that PU.1 functions to repress the IgH intronic enhancer and activate the FcgammaRIIb promoter, while Spi-C opposes these activities. WT-Spi-C pro-B cells have reduced levels of dimethylation on lysine 9 of histone H3 within the IgH 3' regulatory region, indicating that Spi-C can contribute to removal of repressive features in the IgH locus. Overall, these studies suggest that Spi-C may promote B cell differentiation by modulating the activity of PU.1-dependent genes.
Collapse
Affiliation(s)
- Brock L Schweitzer
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | | | | | | | | | | |
Collapse
|
12
|
Mueller BU, Pabst T, Hauser P, Gilliland G, Neuberg D, Tenen DG. Mutations of the transcription factor PU.1 are not associated with acute lymphoblastic leukaemia. Br J Cancer 2006; 94:1918-20. [PMID: 16735999 PMCID: PMC2361337 DOI: 10.1038/sj.bjc.6603198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The transcription factor PU.1 plays a crucial role during normal haematopoiesis in both myeloid cells and B-lymphocytes. Mice with a disruption in both alleles of the PU.1 locus were found to lack macrophages and B cells and had delayed appearance of neutrophils. In addition, critical decrease of PU.1 expression is sufficient to cause acute myeloid leukaemia (AML) and lymphomas in mice. Recently, we reported that heterozygous mutations in the PU.1 gene are present in some patients with AML. Thus, we hypothesised that PU.1 mutations might also contribute to the development of acute leukaemias of the B-cell lineage. Here, we screened 62 patients with B-cell acute lymphoblastic leukaemia (B-ALL) at diagnosis for genomic mutations by direct sequencing of all five exons of the PU.1 gene. We found no genomic alteration of the PU.1 gene suggesting that PU.1 mutations are not likely to be common in B-ALL.
Collapse
Affiliation(s)
- B U Mueller
- Department of Internal Medicine, University Hospital, Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
13
|
Cho SJ, Kang CJ. A Stat5-overlapping site is critical for the IgJ enhancer activity in the plasma cells and bound by a ubiquitous protein. Biochem Biophys Res Commun 2005; 338:1897-905. [PMID: 16288984 DOI: 10.1016/j.bbrc.2005.10.167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 10/26/2005] [Indexed: 10/25/2022]
Abstract
Although the IgJ enhancer chromatin is induced open by an IL-2/Stat5 signaling during terminal B cell differentiation, the opened chromatin of IgJ enhancer is then maintained in the absence of IL-2/Stat5 signaling. Nevertheless, the sequence overlapping the Stat5 site was shown still to be essential for the function of IgJ enhancer in the plasma cells. An in vivo footprint was identified over the Stat5-overlapping site, indicating that the site should be bound by a certain other protein than Stat5. In EMSA using the Stat5-overlapping sequence as a probe, its specific binding protein was identified. The specific binding protein corresponded neither to any of other Stat family proteins, nor to any of potential candidate proteins as tested in EMSA using their corresponding oligo DNA competitors and antibodies. Although its identity remains to be found by its purification, the protein binding specifically to the Stat5-overlapping site was shown to be expressed rather ubiquitously in B and non-B cells, and its molecular weight appeared to be below 52 kDa as determined in the UV-crosslinking-coupled SDS-PAGE.
Collapse
Affiliation(s)
- Sun-Jung Cho
- Graduate School of Biotechnology, Institute of Life Science and Resources, Kyung Hee University, 1 Seocheon-ri, Giheung, Yongin, Gyeonggi-do 449-701, Republic of Korea
| | | |
Collapse
|
14
|
Brandtzaeg P, Johansen FE. Mucosal B cells: phenotypic characteristics, transcriptional regulation, and homing properties. Immunol Rev 2005; 206:32-63. [PMID: 16048541 DOI: 10.1111/j.0105-2896.2005.00283.x] [Citation(s) in RCA: 286] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mucosal antibody defense depends on a complex cooperation between local B cells and secretory epithelia. Mucosa-associated lymphoid tissue gives rise to B cells with striking J-chain expression that are seeded to secretory effector sites. Such preferential homing constitutes the biological basis for local production of polymeric immunoglobulin A (pIgA) and pentameric IgM with high affinity to the epithelial pIg receptor that readily can export these antibodies to the mucosal surface. This ultimate functional goal of mucosal B-cell differentiation appears to explain why the J chain is also expressed by IgG- and IgD-producing plasma cells (PCs) occurring at secretory tissue sites; these immunocytes may be considered as 'spin-offs' from early effector clones that through class switch are on their way to pIgA production. Abundant evidence supports the notion that intestinal PCs are largely derived from B cells initially activated in gut-associated lymphoid tissue (GALT). Nevertheless, insufficient knowledge exists concerning the relative importance of M cells, major histocompatibility complex class II-expressing epithelial cells, and professional antigen-presenting cells for the uptake, processing, and presentation of luminal antigens in GALT to accomplish the extensive and sustained priming and expansion of mucosal B cells. Likewise, it is unclear how the germinal center reaction in GALT so strikingly can promote class switch to IgA and expression of J chain. Although B-cell migration from GALT to the intestinal lamina propria is guided by rather well-defined adhesion molecules and chemokines/chemokine receptors, the cues directing preferential homing to different segments of the gut require better definition. This is even more so for the molecules involved in homing of mucosal B cells to secretory effector sites beyond the gut, and in this respect, the role of Waldever's ring (including the palatine tonsils and adenoids) as a regional inductive tissue needs further characterization. Data suggest a remarkable compartmentalization of the mucosal immune system that must be taken into account in the development of effective local vaccines to protect specifically the airways, eyes, oral cavity, small and large intestines, and urogenital tract.
Collapse
Affiliation(s)
- Per Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Institute and Department of Pathology, University of Oslo, Rikshospitalet University Hospital, Oslo, Norway.
| | | |
Collapse
|
15
|
Listman JA, Wara-aswapati N, Race JE, Blystone LW, Walker-Kopp N, Yang Z, Auron PE. Conserved ETS domain arginines mediate DNA binding, nuclear localization, and a novel mode of bZIP interaction. J Biol Chem 2005; 280:41421-8. [PMID: 16223730 DOI: 10.1074/jbc.m509143200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The DNA-binding ETS transcription factor Spi-1/PU.1 is of central importance in determining the myeloid-erythroid developmental switch and is required for monocyte and osteoclast differentiation. Many monocyte genes are dependent upon this factor, including the gene that codes for interleukin-1beta. It has long been known that the conserved ETS DNA-binding domain of Spi-1/PU.1 functionally cooperates via direct association with a diverse collection of DNA-binding proteins, including members of the basic leucine zipper domain (bZIP) family. However, the molecular basis for this interaction has long been elusive. Using a combination of approaches, we have mapped a single residue on the surface of the ETS domain critical for protein tethering by the C/EBPbeta carboxyl-terminal bZIP domain. This residue is also important for nuclear localization and DNA binding. In addition, dependence upon the leucine zipper suggests a novel mode for both protein-DNA interaction and functional cooperativity.
Collapse
Affiliation(s)
- James A Listman
- SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Polli M, Dakic A, Light A, Wu L, Tarlinton DM, Nutt SL. The development of functional B lymphocytes in conditional PU.1 knock-out mice. Blood 2005; 106:2083-90. [PMID: 15933053 DOI: 10.1182/blood-2005-01-0283] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
An abundance of research has entrenched the view that the Ets domain containing transcription factor PU.1 is fundamental to the development and function of B lymphocytes. In this study, we have made use of a conditional PU.1 allele to test this notion. Complete deletion of PU.1 resulted in the loss of B cells and all other lineage-positive cells in the fetal liver and death between E18.5 and birth; however, specific deletion of PU.1 in the B lineage had no effect on B-cell development. Furthermore, deletion of PU.1 in B cells did not compromise their ability to establish and maintain an immune response. An increased level of apoptosis was observed in vitro upon B-cell receptor (BCR) cross-linking; however, this was partially rescued by interleukin-4 (IL-4). These findings suggest that PU.1 is not essential for the development of functional B lymphocytes beyond the pre-B stage. (Blood. 2005;106:2083-2090)
Collapse
Affiliation(s)
- Matthew Polli
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, Australia 3050
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
The developmental program that commits a hematopoietic stem cell to the B lymphocyte lineage employs transcriptional regulators to enable the assembly of an antigen receptor complex with a useful specificity and with signalling competence. Once a naive IgM+ B cell is generated, it must correctly integrate signals from the antigen receptor with those from cytokine receptors and co-receptors delivering T cell help. The B cell responds through the regulated expression of genes that implement specific cell expansion and differentiation, secretion of high levels of high-affinity antibody, and generation of long-term memory. The transcriptional regulators highlighted in this chapter are those for which genetic evidence of function in IgM+ B cells in vivo has been provided, often in the form of mutant mice generated by conventional or conditional gene targeting. A critical developmental step is the maturation of bone marrow emigrant "transitional" B cells into the mature, long-lived cells of the periphery, and a number of the transcription factors discussed here impact on this process, yielding B cells with poor mitogenic responses in vitro. For mature B cells, it is clear that not only the nature, but the duration and amplitude of an activating signal are major determinants of the transcription factor activities enlisted, and so the ultimate outcome. The current challenge is the identification of the target genes that are activated to implement the correct response, so that we may more precisely and safely manipulate B cell behavior to predictably and positively influence humoral immune responses.
Collapse
Affiliation(s)
- L M Corcoran
- The Walter and Eliza Hall Institute of Medical Research, Victoria, Australia.
| |
Collapse
|
18
|
Garrett-Sinha LA, Hou P, Wang D, Grabiner B, Araujo E, Rao S, Yun TJ, Clark EA, Simon MC, Clark MR. Spi-1 and Spi-B control the expression of the Grap2 gene in B cells. Gene 2005; 353:134-46. [PMID: 15936902 DOI: 10.1016/j.gene.2005.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2004] [Revised: 03/21/2005] [Accepted: 04/07/2005] [Indexed: 11/16/2022]
Abstract
The Ets family members Spi-1 and Spi-B have been implicated in the regulation of genes important for B cell antigen receptor (BCR) signaling. Mice deficient in Spi-B exhibit reduced B cell proliferation in response to BCR cross-linking and impaired T cell-dependent immune responses. This defect is exacerbated in the presence of Spi-1 haplo-insufficiency (Spi1+/- SpiB-/-). Tyrosine phosphorylation and calcium mobilization induced by BCR engagement is diminished in Spi1+/- SpiB-/- B lymphocytes, although many key BCR signaling proteins are expressed, suggesting that Spi-1 and Spi-B regulate expression of additional, unidentified signaling molecules. We now demonstrate that expression of the adaptor protein Grap2 is impaired in Spi1+/- SpiB+/- and Spi1+/- SpiB-/- B lymphocytes. Analysis of two alternate murine Grap2 promoters revealed a functionally important Spi-1 and Spi-B DNA binding element located in the downstream promoter. Ectopic expression of Grap2 in Grap2-deficient B cells reduced the recruitment of BLNK to Igalpha and the phosphorylation of specific substrates. Regulation of BLNK recruitment was dependent upon the Grap2 proline-rich domain, while modulation of phosphorylation was dependent upon both the proline-rich and SH2 domains. These data indicate that Spi-1 and Spi-B directly regulate the expression of Grap2 and that Grap2 functions to modulate BCR signaling, but that reduced Grap2 expression is unlikely to account for the BCR signaling defects observed in Spi1+/- SpiB-/- B cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- B-Lymphocytes/metabolism
- Base Sequence
- Binding Sites/genetics
- Blotting, Northern
- Blotting, Western
- Cell Line, Tumor
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Down-Regulation/genetics
- Electrophoretic Mobility Shift Assay
- Gene Expression/genetics
- Genotype
- Heterozygote
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Mutation
- Oligonucleotides/genetics
- Oligonucleotides/metabolism
- Promoter Regions, Genetic/genetics
- Protein Binding
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Sequence Alignment
- Sequence Homology, Nucleic Acid
- Signal Transduction
- Spleen/cytology
- Spleen/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Lee Ann Garrett-Sinha
- Department of Biochemistry, State University of New York at Buffalo, 140 Farber Hall, 3435 Main Street, Buffalo, NY 14214, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lefebvre JM, Haks MC, Carleton MO, Rhodes M, Sinnathamby G, Simon MC, Eisenlohr LC, Garrett-Sinha LA, Wiest DL. Enforced Expression of Spi-B Reverses T Lineage Commitment and Blocks β-Selection. THE JOURNAL OF IMMUNOLOGY 2005; 174:6184-94. [PMID: 15879115 DOI: 10.4049/jimmunol.174.10.6184] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The molecular changes that restrict multipotent murine thymocytes to the T cell lineage and render them responsive to Ag receptor signals remain poorly understood. In this study, we report our analysis of the role of the Ets transcription factor, Spi-B, in this process. Spi-B expression is acutely induced coincident with T cell lineage commitment at the CD4(-)CD8(-)CD44(-)CD25(+) (DN3) stage of thymocyte development and is then down-regulated as thymocytes respond to pre-TCR signals and develop beyond the beta-selection checkpoint to the CD4(-)CD8(-)CD44(-)CD25(-) (DN4) stage. We found that dysregulation of Spi-B expression in DN3 thymocytes resulted in a dose-dependent perturbation of thymocyte development. Indeed, DN3 thymocytes expressing approximately five times the endogenous level of Spi-B were arrested at the beta-selection checkpoint, due to impaired induction of Egr proteins, which are important molecular effectors of the beta-selection checkpoint. T lineage-committed DN3 thymocytes expressing even higher levels of Spi-B were diverted to the dendritic cell lineage. Thus, we demonstrate that the prescribed modulation of Spi-B expression is important for T lineage commitment and differentiation beyond the beta-selection checkpoint; and we provide insight into the mechanism underlying perturbation of development when that expression pattern is disrupted.
Collapse
Affiliation(s)
- Juliette M Lefebvre
- Immunobiology Working Group, Division of Basic Sciences, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gangenahalli GU, Gupta P, Saluja D, Verma YK, Kishore V, Chandra R, Sharma RK, Ravindranath T. Stem Cell Fate Specification: Role of Master Regulatory Switch Transcription Factor PU.1 in Differential Hematopoiesis. Stem Cells Dev 2005; 14:140-52. [PMID: 15910240 DOI: 10.1089/scd.2005.14.140] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PU.1 is a versatile hematopoietic cell-specific ETS-family transcriptional regulator required for the development of both the inborn and the adaptive immunity, owing to its potential ability to regulate the expression of multiple genes specific for different lineages during normal hematopoiesis. It functions in a cell-autonomous manner to control the proliferation and differentiation, predominantly of lymphomyeloid progenitors, by binding to the promoters of many myeloid genes including the macrophage colony-stimulating factor (M-CSF) receptor, granulocyte-macrophage (GM)-CSF receptor alpha, and CD11b. In B cells, it regulates the immunoglobulin lambda 2-4 and kappa 3' enhancers, and J chain promoters. Besides lineage development, PU.1 also directs homing and long-term engraftment of hematopoietic progenitors to the bone marrow. PU.1 gene disruption causes a cell-intrinsic defect in hematopoietic progenitor cells, recognized by an aberrant myeloid and B lymphoid development. It also immortalizes erythroblasts when overexpressed in many cell lines. Although a number of reviews have been published on its functional significance, in the following review we attempted to consolidate information about the differential participation and role of transcription factor PU.1 at various stages of hematopoietic development beginning from stem cell proliferation, lineage commitment and terminal differentiation into distinct blood cell types, and leukemogenesis.
Collapse
Affiliation(s)
- Gurudutta U Gangenahalli
- Stem Cell Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi-110054, India.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hjortoe GM, Weilguny D, Willumsen BM. Elk3 from hamster--a ternary complex factor with strong transcriptional repressor activity. DNA Cell Biol 2005; 24:35-42. [PMID: 15684718 DOI: 10.1089/dna.2005.24.35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Elk3 belongs to the Ets family of transcription factors, which are regulated by the Ras/mitogen-activated protein kinase-signaling pathway. In the absence of Ras, this protein is a strong inhibitor of transcription and may be directly involved in regulation of growth by downregulating the transcription of genes that are activated during entry into G1. We have isolated the Cricetulus griseus Elk3 gene from the Chinese hamster ovary (CHO) cell line and investigated the transcriptional potential of this factor. Transient transfections revealed that, in addition to its regulation of the c-fos promoter, Elk3 from CHO cells seems to inhibit other promoters controlling expression of proteins involved in G1/S phase progression; Cyclin D1 and DHFR. As has been described for the Elk3 homologs Net (Mouse) and Sap-2 (Human), the results of the present study further indicate that hamster Elk3 is a target of the Ras-Raf-MAPK pathway, and cotransfections with constitutively active H-ras relieves its negative transcriptional activity. No cells stably expressing exogenous Elk3 could be obtained, possibly due to an unspecified toxic or growth retarding effect. These findings support a possible role for Elk3 in growth regulation and reveal a high degree of homology for this protein across species.
Collapse
Affiliation(s)
- Gertrud Malene Hjortoe
- Department Molecular Cell Biology, Institute of Molecular Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
22
|
Organization and Expression of Genes Encoding IgA Heavy Chain, Polymeric Ig Receptor, and J Chain. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
23
|
Torlakovic E, Malecka A, Myklebust JH, Tierens A, Aasheim HC, Nesland JM, Smeland E, Kvaløy S, Delabie J. PU.1 protein expression has a positive linear association with protein expression of germinal centre B cell genes includingBCL-6, CD10, CD20 andCD22: identification of PU.1 putative binding sites in theBCL-6 promotor. J Pathol 2005; 206:312-9. [PMID: 15892171 DOI: 10.1002/path.1777] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The transcription factor PU.1 has been shown to be crucial for the early stages of B cell development but its function at later stages of B cell development is less well known. We observed previously that PU.1 is expressed uniformly throughout the mature pre-plasma cell B cell population, the only exception being a subpopulation of germinal centre (GC) cells which showed exceptionally high expression of PU.1. This suggested that PU.1 may also have a role in GC B cell biology. To test this hypothesis and to screen for possible genes regulated by PU.1, we first evaluated semi-quantitatively the possible co-expression of PU.1 with proteins known to be upregulated or downregulated during GC B cell development. Normal lymphoid tissues and 255 B cell non-Hodgkin lymphomas of putative GC B cell origin were evaluated. PU.1 expression was positively associated with CD10 (p < 0.0001), CD20 (p = 0.043), CD22 (p = 0.005), CD79a (p = 0.024) and Bcl-6 (p < 0.0001) and negatively associated with cytoplasmic immunoglobulin light-chain expression (p = 0.036) in diffuse large B cell lymphoma. Identical or nearly identical associations were found in follicular lymphoma. Since CD20 is known to be partly regulated by PU.1 and putative PU.1-binding sites have been described in the regulatory regions of the CD22, CD79a and CD10 genes, we looked for putative PU.1 binding sites in the BCL6 promotor. Four such putative PU.1 binding sites were identified. Further analysis by gel-shift electromobility essay showed that PU.1 protein binds to three of the four putative binding sites in the BCL6 promotor. PU.1 and Bcl-6 were also found to be upregulated in centroblasts in the normal GC, but jointly downregulated in a subpopulation of centrocytes. Our findings support the contention that PU.1 may also have an important role in GC B cell development.
Collapse
Affiliation(s)
- Emina Torlakovic
- Department of Pathology, The Norwegian Radium Hospital, University of Oslo, Oslo, Norway.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Johansen FE, Brandtzaeg P. Transcriptional regulation of the mucosal IgA system. Trends Immunol 2004; 25:150-7. [PMID: 15036043 DOI: 10.1016/j.it.2004.01.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Finn-Eirik Johansen
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), Institute and Department of Pathology, Rikshospitalet University Hospital, N-0027 Oslo, Norway.
| | | |
Collapse
|
25
|
Schweitzer BL, DeKoter RP. Analysis of Gene Expression and Ig Transcription in PU.1/Spi-B-Deficient Progenitor B Cell Lines. THE JOURNAL OF IMMUNOLOGY 2003; 172:144-54. [PMID: 14688320 DOI: 10.4049/jimmunol.172.1.144] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A number of presumptive target genes for the Ets-family transcription factor PU.1 have been identified in the B cell lineage. However, the precise function of PU.1 in B cells has not been studied because targeted null mutation of the PU.1 gene results in a block to lymphomyeloid development at an early developmental stage. In this study, we take advantage of recently developed PU.1(-/-)Spi-B(-/-) IL-7 and stromal cell-dependent progenitor B (pro-B) cell lines to analyze the function of PU.1 and Spi-B in B cell development. We show that contrary to previously published expectations, PU.1 and/or Spi-B are not required for Ig H chain (IgH) gene transcription in pro-B cells. In fact, PU.1(-/-)Spi-B(-/-) pro-B cells have increased levels of IgH transcription compared with wild-type pro-B cells. In addition, high levels of Igkappa transcription are induced after IL-7 withdrawal of wild-type or PU.1(-/-)Spi-B(-/-) pro-B cells. In contrast, we found that Iglambda transcription is reduced in PU.1(-/-)Spi-B(-/-) pro-B cells relative to wild-type pro-B cells after IL-7 withdrawal. These results suggest that Iglambda, but not IgH or Igkappa, transcription, is dependent on PU.1 and/or Spi-B. The PU.1(-/-)Spi-B(-/-) pro-B cells have other phenotypic changes relative to wild-type pro-B cells including increased proliferation, increased CD25 expression, decreased c-Kit expression, and decreased RAG-1 expression. Taken together, our observations suggest that reduction of PU.1 and/or Spi-B activity in pro-B cells promotes their differentiation to a stage intermediate between late pro-B cells and large pre-B cells.
Collapse
Affiliation(s)
- Brock L Schweitzer
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, Medical Sciences Building 3006, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | | |
Collapse
|
26
|
Salas M, Eckhardt LA. Critical Role for the Oct-2/OCA-B Partnership in Ig-Secreting Cells. THE JOURNAL OF IMMUNOLOGY 2003; 171:6589-98. [PMID: 14662861 DOI: 10.4049/jimmunol.171.12.6589] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B and T lymphocytes arise from a common precursor in the bone marrow, but ultimately acquire very different functions. The difference in function is largely attributable to the expression of tissue-specific transcription factors that activate discrete sets of genes. In previous studies we and others have shown that the specialized genes expressed by Ig-secreting cells cease transcription when these cells are fused to a T lymphoma. The extinguished genes include those encoding Ig, J chain, and the transcription factors Oct-2, PU.1, and the coactivator OCA-B. Remarkably, if we sustain Oct-2 expression during cell fusion, all the other tissue-specific genes of the Ig-secreting cell simultaneously escape silencing. This suggests that Oct-2 plays a central role in maintaining the gene expression program of these cells. In the present studies we have investigated the roles of the transcription factor PU.1 and the coactivator OCA-B within the hierarchy of regulatory factors that sustain Ig-secreting cell function. Our results show that OCA-B and Oct-2 are regulatory partners in this process and that PU.1 plays a subordinate role at this cell stage.
Collapse
Affiliation(s)
- Mabel Salas
- Department of Biological Sciences, Hunter College and Graduate Center of City University of New York, New York, NY 10021, USA
| | | |
Collapse
|
27
|
Dekoninck A, Calomme C, Nizet S, de Launoit Y, Burny A, Ghysdael J, Van Lint C. Identification and characterization of a PU.1/Spi-B binding site in the bovine leukemia virus long terminal repeat. Oncogene 2003; 22:2882-96. [PMID: 12771939 DOI: 10.1038/sj.onc.1206392] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bovine leukemia virus (BLV) is a B-lymphotropic oncogenic retrovirus whose transcriptional promoter is located in the viral 5' long terminal repeat (LTR). To date, no B-lymphocyte-specific cis-regulatory element has been identified in this region. Since ETS proteins are known to regulate transcription of numerous retroviruses, we searched for the presence in the BLV promoter region of binding sites for PU.1/Spi-1, a B-cell- and macrophage-specific ETS family member. In this report, nucleotide sequence analysis of the viral LTR identified a PUbox located at -95/-84 bp. We demonstrated by gel shift and supershift assays that PU.1 and the related Ets transcription factor Spi-B interacted specifically with this PUbox. A 2-bp mutation (GGAA-->CCAA) within this motif abrogated PU.1/Spi-B binding. This mutation caused a marked decrease in LTR-driven basal gene expression in transient transfection assays of B-lymphoid cell lines, but did not impair the responsiveness of the BLV promoter to the virus-encoded transactivator Tax(BLV). Moreover, ectopically expressed PU.1 and Spi-B proteins transactivated the BLV promoter in a PUbox-dependent manner. Taken together, our results provide the first demonstration of regulation of the BLV promoter by two B-cell-specific Ets transcription factors, PU.1 and Spi-B. The PU.1/Spi-B binding site identified here could play an important role in BLV replication and B-lymphoid tropism.
Collapse
Affiliation(s)
- Ann Dekoninck
- Laboratoire de Virologie Moléculaire, Service de Chimie Biologique, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, Rue des Profs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | | | | | | | | | | | | |
Collapse
|
28
|
Wang JM, Lai MZ, Yang-Yen HF. Interleukin-3 stimulation of mcl-1 gene transcription involves activation of the PU.1 transcription factor through a p38 mitogen-activated protein kinase-dependent pathway. Mol Cell Biol 2003; 23:1896-909. [PMID: 12612065 PMCID: PMC149468 DOI: 10.1128/mcb.23.6.1896-1909.2003] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have previously demonstrated that the antiapoptotic gene mcl-1 is activated by interleukin-3 (IL-3) in Ba/F3 pro-B cells through two promoter elements designated the CRE-2 and SIE motifs. While the CRE-2-binding complex contains the CREB protein and is activated by IL-3 through the phosphatidylinositol 3-kinase/Akt-dependent pathway, the identity and cytokine activation pathway of the SIE-binding complex remains unclear. In this report, we demonstrated that PU.1 is one component of the SIE-binding complex. A chromatin immunoprecipitation assay further confirmed that PU.1 binds to the mcl-1 promoter region containing the SIE motif in vivo. While IL-3 stimulation does not significantly alter the SIE-binding activity of PU.1, it markedly increases PU.1's transactivation activity. The latter effect coincides with the increased phosphorylation of PU.1 following IL-3 activation of a p38 mitogen-activated protein kinase (p38(MAPK))-dependent pathway. A serine-to-alanine substitution at position 142 significantly weakens PU.1's ability to be phosphorylated by the p38(MAPK) immunocomplex. Furthermore, this S142A mutant is impaired in the ability to be further stimulated by IL-3 to transactivate the mcl-1 reporter through the SIE motif. Taken together, our results demonstrate that IL-3 stimulation of mcl-1 gene transcription through the SIE motif involves phosphorylation of PU.1 at serine 142 by a p38(MAPK)-dependent pathway.
Collapse
Affiliation(s)
- Ju-Ming Wang
- Institute of Molecular Biology, Academia Sinica, 128 Yen-Jiou Yuan Road, Section 2, Nankang, Taipei 11529, Taiwan, Republic of China
| | | | | |
Collapse
|
29
|
Arguello M, Sgarbanti M, Hernandez E, Mamane Y, Sharma S, Servant M, Lin R, Hiscott J. Disruption of the B-cell specific transcriptional program in HHV-8 associated primary effusion lymphoma cell lines. Oncogene 2003; 22:964-73. [PMID: 12592383 DOI: 10.1038/sj.onc.1206270] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Primary effusion lymphoma (PEL) is a lymphoproliferative disease of B-cell origin that is associated with HHV-8 infection. PEL cells harbor a non-B, non-T phenotype and lack significant surface immunoglobulin (Ig) expression, a characteristic that has not been fully explained. In the present study, we demonstrate that PEL cells constitutively express interferon regulatory factor (IRF)-4, a transcription factor that regulates the activity of the immunoglobulin light-chain enhancer elements lambdaB and kappaE3' through binding to a composite Ets-IRF site. IRF-4 activity requires its physical interaction with PU.1, an Ets family member involved in the activation of genes essential for B-cell development. However, in PEL-derived B-cell lines, PU.1 expression was completely abrogated; expression of the B cell specific transcription factor Oct-2, which is known to regulate PU.1 expression, was also abolished. Moreover, the B-cell-specific coactivator of octamer factors, BOB-1/OcaB, was expressed at very decreased levels in PEL cells. Ectopic expression of Oct-2 was able to fully restore PU.1 promoter activity in the PEL cell line BCBL-1, while PU.1 expression also reconstituted the activity of the lambdaB Ets-IRF site. In addition, protein levels of BSAP/Pax-5 and IRF-8/ICSBP were undetectable in PEL cells. The pattern of transcription factor ablation observed in PEL was found to be comparable to that observed in classical Hodgkin's disease-derived cell lines, which also lack B-cell-specific surface markers. These observations indicate that disruption of the B-cell-specific transcriptional program is likely to contribute to the incomplete B-cell phenotype characteristic of PEL cells.
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- Body Fluids
- Burkitt Lymphoma/genetics
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/pathology
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Enhancer Elements, Genetic
- Gene Expression Regulation, Neoplastic
- Herpesviridae Infections/genetics
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/pathology
- Herpesvirus 8, Human/isolation & purification
- Hodgkin Disease/genetics
- Hodgkin Disease/metabolism
- Hodgkin Disease/pathology
- Humans
- Immunoglobulin Light Chains/genetics
- Interferon Regulatory Factors
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/virology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Octamer Transcription Factor-2
- PAX5 Transcription Factor
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- RNA, Messenger/biosynthesis
- RNA, Neoplasm/biosynthesis
- Recombinant Fusion Proteins/physiology
- Repressor Proteins/biosynthesis
- Repressor Proteins/genetics
- Trans-Activators/biosynthesis
- Trans-Activators/genetics
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Transcription, Genetic
- Transfection
- Tumor Virus Infections/genetics
- Tumor Virus Infections/metabolism
- Tumor Virus Infections/pathology
Collapse
Affiliation(s)
- Meztli Arguello
- Terry Fox Molecular Oncology Group, Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Toda Y, Tsukada J, Misago M, Kominato Y, Auron PE, Tanaka Y. Autocrine induction of the human pro-IL-1beta gene promoter by IL-1beta in monocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1984-91. [PMID: 11823535 DOI: 10.4049/jimmunol.168.4.1984] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-1beta is produced primarily by activated monocytes/macrophages. We report in this study that IL-1beta induces the human pro-IL-1beta (IL1B) gene promoter in human THP-1 monocytic cells. The -131 to +12 minimal IL1B promoter was induced by IL-1beta in a dose-dependent manner. The promoter possesses two important transcription factor binding motifs, one for an ETS family transcription factor Spi-1 (PU.1), and the other a binding site for NF-IL6 (CCAAT/enhancer binding protein beta). Autocrine promoter activity was completely inhibited by mutation of the Spi-1 site. Mutation of the NF-IL6 binding motif caused partial loss of activity. EMSAs using THP-1 cell nuclear extracts indicated that IL-1beta significantly induced Spi-1 binding to its target site within the IL1B promoter that was maximal at 1 h after stimulation, correlating with the kinetics of IL-1beta induction. The importance of Spi-1 was supported by our observation that Spi-1-deficient EL4 thymocytes exhibited IL-1beta-induced activity only after transfection with a Spi-1 expression vector. Moreover, TNFR-associated factor 6 also required Spi-1 to activate the promoter. Transfection studies using Spi-1 mutant constructs showed that the TATA-binding protein binding and glutamine-rich domains of Spi-1 were important for IL-1beta induction, whereas LPS induction required the proline, glutamic acid, serine, and threonine-rich domain containing serine 148 as well as the TATA-binding protein and glutamine-rich domains. We conclude that the IL1B promoter is an IL-1beta-responsive sequence as a result of its ability to bind Spi-1 in response to IL-1beta.
Collapse
Affiliation(s)
- Yoko Toda
- First Department of Internal Medicine, School of Medicine, and School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Takahashi T, Kimura M, Matsumoto N, Iwata A, Ogura Y, Yoshida T, Kamei N, Komiyama K, Mestecky J, Moro I. Cloning of the chicken immunoglobulin joining (J)-chain gene and characterization of its promoter region. DNA Cell Biol 2002; 21:81-90. [PMID: 11953007 DOI: 10.1089/104454902753604952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Three overlapping genomic clones of the chicken immunoglobulin joining (J) chain were isolated and then characterized using restriction enzyme analysis, Southern blot analysis with cDNA probes, and DNA sequencing. The gene consisted of four exons separated by a 2.6-kb intron 1, a 0.9-kb intron 2, and a 0.5-kb intron 3. A transcriptional initiation site was identified by a primer extension method using mRNA and cDNA, indicating that exon 1 was 86 bp encoding 20 amino acid residues. A TATA box was positioned at 29 approximately 25 bp upstream of exon 1. Exons, 2, and 3 consisted of 133 bp and 81 bp, encoding 43 and 26 amino acid residues of the mature protein, respectively. Exon 4 consisted of 202 bp encoding 66 amino acid residues and 1.2 kb of untranslated sequence. Deletion mutants of a 4.1-kb genomic fragment containing exon 1 showed high levels of promoter activities when examined in luciferase reporter assays following transfection into the DT-40 chicken B-cell line. These results suggest that the chicken J-chain gene consists of four exons and three introns and that the transcriptional regulatory elements may be present within 3.8 kb upstream of exon 1.
Collapse
Affiliation(s)
- Tomihisa Takahashi
- Department of Pathology, Nihon University School of Dentistry, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
EL-Gazzar MA, Maeda K, Nomiyama H, Nakao M, Kuwahara K, Sakaguchi N. PU.1 is involved in the regulation of B lineage-associated and developmental stage-dependent expression of the germinal center-associated DNA primase GANP. J Biol Chem 2001; 276:48000-8. [PMID: 11641399 DOI: 10.1074/jbc.m106696200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Germinal center-associated DNA primase (GANP) associated with MCM3 of the DNA replication complex is up-regulated selectively in germinal center B cells. We studied promoter activity of the 5' region involved in the developmental stage-dependent expression in B lineage cells by luciferase reporter assay. Selective regulation of ganp expression was observed in the -737-bp promoter region in B and plasma cell lines but was significantly low in pre-B and T cell lines. The deletion constructs displayed a gap decrease after shortening the region from -134 to -108 bp. Further narrowing suggested the involvement of the PU.1 consensus sequence at -126 bp by electrophoretic mobility shift assay. The protein component PU.1 complex is not inhibited with mutated probes at the consensus site but is inhibited with the known PU.1 probe of CD72 and with anti-PU.1 antibody. Moreover, introduction of PU.1 cDNA enhanced the reporter gene activity in a dose-dependent manner in B cells, whereas the reporter construct with the mutated PU.1 site did not respond. Anti-CD40 stimulation induced the reporter activity with a 100% increase, which is not observed with the PU.1-mutated reporter construct. These results demonstrate that the germinal center-associated DNA primase expression is partly regulated by the transcription factor PU.1 expressed in B lineage cells.
Collapse
Affiliation(s)
- M A EL-Gazzar
- Department of Immunology, Kumamoto University School of Medicine, 2-2-1, Honjo, Kumamoto, 860-0811 Japan
| | | | | | | | | | | |
Collapse
|
33
|
Torlakovic E, Tierens A, Dang HD, Delabie J. The transcription factor PU.1, necessary for B-cell development is expressed in lymphocyte predominance, but not classical Hodgkin's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:1807-14. [PMID: 11696441 PMCID: PMC1867045 DOI: 10.1016/s0002-9440(10)63027-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hodgkin's disease (HD) is a lymphoproliferative disease of predominantly B-cell origin. However, the reasons for the incomplete development of the B-cell phenotype and lack of immunoglobulin expression in classical HD (cHD) have not been fully explained. We examined the expression of PU.1 in HD, an Ets-family transcription factor, which regulates the expression of immunoglobulin and other genes that are important for B-cell development. Immunohistochemistry for PU.1 was performed on 35 cases of cHD and 15 cases of lymphocyte predominance HD as well as 67 non-Hodgkin's lymphomas (NHL). Expression of PU.1 was studied by Western blotting in four cHD-derived cell lines and in five NHL cell lines. We also studied the expression of two additional B-cell transcription factors, B-cell-specific activator protein and Oct-2. Our results show a striking lack of PU.1 expression by neoplastic cells in cHD but not in lymphocyte predominance HD. Our study also confirmed that B-cell-specific activator protein but not Oct-2 is not expressed by cHD. Western blotting showed no PU.1 protein expression in the cHD-derived cell lines, with the exception of one cell line of putative monocyte/histiocyte origin. The lack of PU.1 protein expression in cHD likely contributes to the lack of immunoglobulin expression and incomplete B-cell phenotype characteristic of the Reed-Sternberg cells in cHD.
Collapse
Affiliation(s)
- E Torlakovic
- Department of Pathology, The Norwegian Radium Hospital, Oslo, Norway.
| | | | | | | |
Collapse
|
34
|
Hu CJ, Rao S, Ramirez-Bergeron DL, Garrett-Sinha LA, Gerondakis S, Clark MR, Simon MC. PU.1/Spi-B regulation of c-rel is essential for mature B cell survival. Immunity 2001; 15:545-55. [PMID: 11672537 DOI: 10.1016/s1074-7613(01)00219-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PU.1(+/-)Spi-B(-/-) mice exhibit reduced numbers of immature and mature B lymphocytes, which exhibit severe defects in response to BCR-mediated stimulation and poor survival. We found that expression of c-rel, a member of the Rel/NF-kappa B family, is dramatically reduced in PU.1(+/-)Spi-B(-/-) splenic B cells. Analysis of the murine c-rel promoter identified three PU.1/Spi-B binding sites critical for c-rel promoter activity. Furthermore, reintroduction of Rel protein restored wild-type B cell numbers to mice reconstituted with PU.1(+/-)Spi-B(-/-) bone marrow. These findings are the first to demonstrate that a member of the Rel/NF-kappa B family is directly regulated by Ets proteins and dissect the molecular basis for the function of two Ets factors, PU.1 and Spi-B, in promoting B lymphocyte survival.
Collapse
Affiliation(s)
- C J Hu
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Barreda DR, Belosevic M. Transcriptional regulation of hemopoiesis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2001; 25:763-789. [PMID: 11602195 DOI: 10.1016/s0145-305x(01)00035-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The regulation of blood cell formation, or hemopoiesis, is central to the replenishment of mature effector cells of innate and acquired immune responses. These cells fulfil specific roles in the host defense against invading pathogens, and in the maintenance of homeostasis. The development of hemopoietic cells is under stringent control from extracellular and intracellular stimuli that result in the activation of specific downstream signaling cascades. Ultimately, all signal transduction pathways converge at the level of gene expression where positive and negative modulators of transcription interact to delineate the pattern of gene expression and the overall cellular hemopoietic response. Transcription factors, therefore, represent a nodal point of hemopoietic control through the integration of the various signaling pathways and subsequent modulation of the transcriptional machinery. Transcription factors can act both positively and negatively to regulate the expression of a wide range of hemopoiesis-relevant genes including growth factors and their receptors, other transcription factors, as well as various molecules important for the function of developing cells. The expression of these genes is dependent on the complex interactions between transcription factors, co-regulatory molecules, and specific binding sequences on the DNA. Recent advances in various vertebrate and invertebrate systems emphasize the importance of transcription factors for hemopoiesis control and the evolutionary conservation of several of such mechanisms. In this review we outline some of the key issues frequently identified in studies of the transcriptional regulation of hemopoietic gene expression. In teleosts, we expect that the characterization of several of these transcription factors and their regulatory mechanisms will complement recent advances in a number of fish systems where identification of cytokine and other hemopoiesis-relevant factors are currently under investigation.
Collapse
Affiliation(s)
- D R Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2E9
| | | |
Collapse
|
36
|
Rieske P, Pongubala JM. AKT induces transcriptional activity of PU.1 through phosphorylation-mediated modifications within its transactivation domain. J Biol Chem 2001; 276:8460-8. [PMID: 11133986 DOI: 10.1074/jbc.m007482200] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Signal transduction by the antigen receptor complexes is critical for developmental progression of B-lymphocytes, which are defined by assembly and sequential expression of immunoglobulin genes, which in turn are regulated by the enhancer elements. Although proximal antigen-receptor signal transduction pathways are well defined, the precise nuclear factors targeted by these signals remained unknown. Previous studies have demonstrated that tissue-restricted transcription factors including PU.1 and PU.1 interaction partner (PIP) function synergistically with c-Fos plus c-Jun to stimulate the kappaE3'-enhancer in 3T3 cells. In this study, we demonstrate that the functional synergy between these factors is enhanced in response to mitogen-activated protein kinase kinase kinase, in 3T3 cells, where the enhancer is inactive. However in S194 plasmacytoma cells, mitogen-activated protein kinase kinase kinase was able to stimulate the activity of PU.1 but unable to induce the kappaE3'-enhancer activity. We have found that Ras-phosphoinositide 3-kinase-dependent externally regulated kinase, AKT, induces kappaE3'-enhancer activity in both pre-B and plasmacytoma cells. AKT stimulation of the kappaE3'-enhancer is primarily due to PU.1 induction and is independent of PU.1 interaction with PIP. Activation of AKT had no effect on the expression levels of PU.1 or its protein-protein interaction with PIP. Using a series of deletion constructs, we have determined that the PU.1 acid-rich (amino acids 33-74) transactivation domain is necessary for AKT-mediated induction. Substitution analyses within this region indicate that phosphorylation of Ser(41) is necessary to respond to AKT. Consistent with these studies, ligation of antigen receptors in A20 B cells mimics AKT activation of PU.1. Taken together, these results provide evidence that PU.1 is induced by AKT signal in a phosphoinositide 3-kinase-dependent manner, leading to inducible or constitutive activation of its target genes.
Collapse
Affiliation(s)
- P Rieske
- Department of Biochemistry, MCP Hahnemann University School of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | |
Collapse
|
37
|
Linderson Y, French NS, Neurath MF, Pettersson S. Context-dependent Pax-5 repression of a PU.1/NF-kappaB regulated reporter gene in B lineage cells. Gene 2001; 262:107-14. [PMID: 11179673 DOI: 10.1016/s0378-1119(00)00546-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Enhancers located in the 3' end of the locus in part regulate immunoglobulin heavy chain (IgH) gene expression. One of these enhancers, HS 1,2, is developmentally regulated by DNA binding proteins like NF-kappaB, Pax-5 and the protein complex NF-alphaP in B lineage cells. Here we report that NF-alphaP is the ets protein PU.1. A glutathione-S-transferase (GST)-pulldown assay demonstrated that PU.1 can physically interact with NF-kappaB in solution. Experiments in COS cells showed that PU.1 and NF-kappaB (p50/c-Rel) can activate transcription of an enhancer linked reporter gene. The paired domain protein Pax-5 has previously been shown to repress enhancer-dependent transcription. Additional co-transfection experiments revealed that PU.1/NF-kappaB dependent transcription could be repressed in a context dependent manner by Pax-5, but not by the paired domain of Pax-5. When the PU.1 binding site was substituted with a binding site for the ets-protein Elf-1, Pax-5 could no longer repress reporter gene activity. Our data indicate a model where Pax-5 mediated repression of the HS 1,2 enhancer requires the recruitment of a co-factor which is dependent on Pax-5/PU.1 but which cannot be recruited by Pax-5/Elf-1.
Collapse
Affiliation(s)
- Y Linderson
- Center for Genomics Research, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | | | | | |
Collapse
|
38
|
Abstract
Ets is a family of transcription factors present in species ranging from sponges to human. All family members contain an approximately 85 amino acid DNA binding domain, designated the Ets domain. Ets proteins bind to specific purine-rich DNA sequences with a core motif of GGAA/T, and transcriptionally regulate a number of viral and cellular genes. Thus, Ets proteins are an important family of transcription factors that control the expression of genes that are critical for several biological processes, including cellular proliferation, differentiation, development, transformation, and apoptosis. Here, we tabulate genes that are regulated by Ets factors and describe past, present and future strategies for the identification and validation of Ets target genes. Through definition of authentic target genes, we will begin to understand the mechanisms by which Ets factors control normal and abnormal cellular processes.
Collapse
Affiliation(s)
- V I Sementchenko
- Center for Molecular and Structural Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, SC 29403, USA
| | | |
Collapse
|
39
|
Rao S, Garrett-Sinha LA, Yoon J, Simon MC. The Ets factors PU.1 and Spi-B regulate the transcription in vivo of P2Y10, a lymphoid restricted heptahelical receptor. J Biol Chem 1999; 274:34245-52. [PMID: 10567398 DOI: 10.1074/jbc.274.48.34245] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To investigate the in vivo functions of PU.1 and Spi-B, two highly related Ets transcription factors, we previously generated PU. 1(+/+)Spi-B(-/-) and PU.1(+/-)Spi-B(-/-) mice and demonstrated a significant decrease in B-cell receptor (BCR) signaling in mutants. Major components of BCR signaling appear to be expressed at normal levels in these mice, implying that PU.1 and Spi-B cooperate in the transcription of additional target genes important for antigen receptor signaling. We used subtractive hybridization to identify novel in vivo PU.1/Spi-B target genes and determined that the expression of a heptahelical receptor, P2Y10, is dramatically reduced in PU.1(+/-)Spi-B(-/-) B-cells. Further analysis shows that P2Y10 expression is restricted to lymphoid cells and parallels that of Spi-B in B-lymphocytes. Lastly, the P2Y10 promoter contains a PU. 1/Spi-B binding site functionally required for efficient transcription in B-cells. Thus, P2Y10 is likely to be a direct in vivo transcriptional target for PU.1 and Spi-B and provides a unique model to explore transcriptional regulation by this Ets factor subfamily. Furthermore, P2Y10 suggests an intriguing connection between heterotrimeric G-proteins and BCR signaling.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Sequence
- Animals
- B-Lymphocytes/metabolism
- Binding Sites
- Binding, Competitive
- Blotting, Northern
- Cell Lineage
- DNA/genetics
- DNA/metabolism
- DNA, Complementary/genetics
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/physiology
- Gene Expression
- Mice
- Mice, Knockout
- Molecular Sequence Data
- Promoter Regions, Genetic
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- RNA, Messenger/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Sequence Homology, Amino Acid
- Tissue Distribution
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Trans-Activators/physiology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcription, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- S Rao
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
40
|
Brandtzaeg P, Farstad IN, Johansen FE, Morton HC, Norderhaug IN, Yamanaka T. The B-cell system of human mucosae and exocrine glands. Immunol Rev 1999; 171:45-87. [PMID: 10582165 PMCID: PMC7159139 DOI: 10.1111/j.1600-065x.1999.tb01342.x] [Citation(s) in RCA: 205] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mucosae and exocrine glands harbour the largest activated B-cell system of the body, amounting to some 80-90% of all immunoglobulin (Ig)-producing cells. The major product of these immunocytes is polymeric (p)IgA (mainly dimers) with associated J chain. Both pIgA and pentameric IgM contain a binding site for the polymeric Ig receptor (pIgR), or secretory component (SC), which is a requirement for their active external transport through secretory epithelia. The pIgR/SC binding site depends on covalent incorporation of the J chain into the quaternary structure of the polymers when they are produced by the local immunocytes. This important differentiation characteristic appears to be sufficient functional justification for the J chain to be expressed also by most B cells terminating at secretory effector sites with IgD or IgG production; they probably represent a "spin-off" from sequential downstream CH switching on its way to pIgA expression, thus apparently reflecting a maturational stage of effector B-cell clones compatible with homing to these sites. Observations in IgA-deficient individuals suggest that the magnitude of this homing is fairly well maintained even when the differentiation pathway to IgA is blocked. Certain microenvironmental elements such as specific cytokines and dendritic cells appear to be required for induction of IgA synthesis, but it remains virtually unknown why this isotype normally is such a dominating product of local immunocytes and why they have such a high level of J chain expression. Also, despite the recent identification of some important requirements in terms of adhesion molecules (e.g. integrin alpha 4 beta 7 and MAdCAM-1) that explain the "gut-seeking" properties of enterically induced B cells, the origin of regionalized homing of B cells to secretory effector sites outside the gut remains elusive. Moreover, little is known about immune regulation underlying the striking disparity of both the class (IgD, IgM) and subclass (IgA1, IgA2, IgG1, IgG2) production patterns shown by local immunocytes in various regions of the body, although the topical microbiota and other environmental stimuli might be important. Rational design of local vaccines will depend on better knowledge of both inductive and migratory properties of human mucosal B cells.
Collapse
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), University of Oslo, National Hospital, Norway.
| | | | | | | | | | | |
Collapse
|
41
|
Pio F, Assa-Munt N, Yguerabide J, Maki RA. Mutants of ETS domain PU.1 and GGAA/T recognition: free energies and kinetics. Protein Sci 1999; 8:2098-109. [PMID: 10548056 PMCID: PMC2144130 DOI: 10.1110/ps.8.10.2098] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The ETS family members display specific DNA binding site preferences. As an example, PU.1 and ETS-1 recognize different DNA sequences with a core element centered over 5'-GGAA-3' and 5'-GGAA/T-3', respectively. To understand the molecular basis of this recognition, we carried out site-directed mutagenesis experiments followed by DNA binding studies that use electrophoretic mobility shift assay (EMSA) and surface plasmon resonance methods. EMSA experiments identified amino acid changes A231S and/or N236Y as being important for PU.1 recognition of both 5'-GGAA-3' and 5'-GGAT-3' containing oligonucleotides. To confirm these data and obtain accurate binding parameters, we performed kinetic studies using surface plasmon resonance on these mutants. The N236Y substitution revealed a weak protein-DNA interaction with the 5'-GGAA-3' containing oligonucleotide caused by a faster release of the protein from the DNA (k(off) tenfold higher than the wild-type protein). With the double mutant A231S-N236Y, we obtained an increase in binding affinity and stability toward both 5'-GGAA-3' and 5'-GGAT-3' containing oligonucleotides. We propose that substitution of alanine for serine introduces an oxygen atom that can accept hydrogen and interact with potential water molecules or other atoms to make an energetically favorable hydrogen bond with both 5'-GGAA-3' and 5'-GGAT-3' oligonucleotides. The free energy of dissociation for the double mutant A231S-N236Y with 5'-GGAA-3' (delta deltaG((A231S-N236Y) - (N236Y)) = -1.2 kcal mol confirm the stabilizing effect of this mutant in the protein-DNA complex formation. We conclude that N236Y mutation relaxes the specificity toward 5'-GGAA-3' and 5'-GGAT-3' sequences, while A231S mutation modulates the degree of specificity toward 5'-GGAA-3' and 5'GGAT-3' sequences. This study explains why wild-type PU.1 does not recognize 5'-GGAT-3' sequences and in addition broadens our understanding of 5'-GGAA/T-3' recognition by ETS protein family members.
Collapse
Affiliation(s)
- F Pio
- The Burnham Institute, La Jolla, California 92037, USA.
| | | | | | | |
Collapse
|
42
|
Wallin JJ, Rinkenberger JL, Rao S, Gackstetter ER, Koshland ME, Zwollo P. B cell-specific activator protein prevents two activator factors from binding to the immunoglobulin J chain promoter until the antigen-driven stages of B cell development. J Biol Chem 1999; 274:15959-65. [PMID: 10336503 DOI: 10.1074/jbc.274.22.15959] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The immunoglobulin J chain gene is inducibly transcribed in mature B cells upon antigen recognition and a signal from interleukin-2 (IL-2). B cell-specific activator protein (BSAP), a transcription factor that silences J chain transcription, has been identified as a nuclear target of the IL-2 signal. The levels of BSAP progressively decrease in response to IL-2 and this change correlates with the differentiation of B cells into antibody secreting plasma cells. Here we report the binding of the upstream stimulatory factor (USF) to an E-box motif immediately upstream from the BSAP site on the J chain promoter. Mutations in the USF binding motif significantly decrease J chain promoter activity in J chain expressing B cell lines. We also show that a functional relationship exists between USF and a second J chain positive-regulating factor, B-MEF2, using co-immunoprecipitation assays and transfections. Finally, we provide evidence that the binding of BSAP prevents USF and B-MEF2 from interacting with the J chain promoter during the antigen-independent stages of B cell development. It is not until the levels of BSAP decrease during the antigen-driven stages of B cell development that both USF and B-MEF2 are able to bind to their respective promoter elements and activate J chain transcription.
Collapse
Affiliation(s)
- J J Wallin
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA
| | | | | | | | | | | |
Collapse
|
43
|
Rao S, Matsumura A, Yoon J, Simon MC. SPI-B activates transcription via a unique proline, serine, and threonine domain and exhibits DNA binding affinity differences from PU.1. J Biol Chem 1999; 274:11115-24. [PMID: 10196196 DOI: 10.1074/jbc.274.16.11115] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SPI-B is a B lymphocyte-specific Ets transcription factor that shares a high degree of similarity with PU.1/SPI-1. In direct contrast to PU.1(-/-) mice that die in utero and lack monocytes, neutrophils, B cells, and T cells, Spi-B-/- mice are viable and exhibit a severe B cell proliferation defect. Since PU.1 is expressed at wild type levels in Spi-B-/- B cells, the mutant mice provide genetic evidence that SPI-B and PU.1 have at least some non-redundant roles in B lymphocytes. To begin to understand the molecular basis for these defects, we delineated functional domains of SPI-B for comparison to those of PU.1. By using a heterologous co-transfection system, we identified two independent transactivation domains in the N terminus of SPI-B. Interestingly, only one of these domains (amino acids 31-61), a proline/serine/threonine-rich region, unique among Ets proteins, is necessary for transactivation of the immunoglobulin lambda light chain enhancer. This transactivation motif is in marked contrast to PU.1, which contains acidic and glutamine-rich domains. In addition, we describe a functional PU.1 site within the c-FES promoter which SPI-B fails to bind efficiently and transactivate. Finally, we show that SPI-B interacts with the PU.1 cofactors Pip, TBP, c-Jun and with lower affinity to nuclear factor interleukin-6beta and retinoblastoma. Taken together, these data suggest that SPI-B binds DNA with a different affinity for certain sites than PU.1 and harbors different transactivation domains. We conclude that SPI-B may activate unique target genes in B lymphocytes and interact with unique, although currently unidentified, cofactors.
Collapse
Affiliation(s)
- S Rao
- Department of Pathology, the University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
44
|
Bemark M, Mårtensson A, Liberg D, Leanderson T. Spi-C, a novel Ets protein that is temporally regulated during B lymphocyte development. J Biol Chem 1999; 274:10259-67. [PMID: 10187812 DOI: 10.1074/jbc.274.15.10259] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A novel Ets protein was isolated by yeast one-hybrid screening of a cDNA library made from lipopolysaccharide-stimulated mouse splenic B cells, using the SP6 kappa promoter kappaY element as a bait. The novel Ets protein was most closely related to PU.1 and Spi-B within the DNA binding Ets domain and was therefore named Spi-C. However, Spi-C may represent a novel subgroup within the Ets protein family, as it differed significantly from Spi-B and PU.1 within helix 1 of the Ets domain. Spi-C was encoded by a single-copy gene that was mapped to chromosome 10, region C. Spi-C interacted with DNA similarly to PU.1 as judged by methylation interference, band-shift and site selection analysis, and activated transcription of a kappaY element reporter gene upon co-transfection of HeLa cells. Spi-C RNA was expressed in mature B lymphocytes and at lower levels in macrophages. Furthermore, pre-B cell and plasma cell lines were Spi-C-negative, suggesting that Spi-C might be a regulatory molecule during a specific phase of B lymphoid development.
Collapse
Affiliation(s)
- M Bemark
- Immunology Unit, Department of Cell and Molecular Biology, Lund University, P. O. Box 7031, S-220 07 Lund, Sweden
| | | | | | | |
Collapse
|
45
|
Garrett-Sinha LA, Su GH, Rao S, Kabak S, Hao Z, Clark MR, Simon MC. PU.1 and Spi-B are required for normal B cell receptor-mediated signal transduction. Immunity 1999; 10:399-408. [PMID: 10229183 DOI: 10.1016/s1074-7613(00)80040-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PU.1 and Spi-B have previously been implicated in the regulation of genes encoding B cell receptor (BCR) signaling components. Spi-B-/- B lymphocytes respond poorly to BCR stimulation; PU.1-/- mice, however, lack B cells, precluding an analysis of BCR responses. We now show that PU.1+/- Spi-B-/- B cells exhibit more extensive defects than Spi-B-/- B cells, indicating that both PU.1 and Spi-B are required for normal BCR signaling. Strikingly, BCR cross-linking results in substantially reduced protein tyrosine phosphorylation in mutant B cells. Further analysis shows that Igalpha is phosphorylated and syk is recruited and becomes phosphorylated but that BLNK and PLCgamma phosphorylation are defective in mutant cells. Our data support the existence of a novel component coupling syk to downstream targets.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Calcium Signaling/immunology
- Cell Lineage/genetics
- Cell Lineage/immunology
- Crosses, Genetic
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/physiology
- Interferon Regulatory Factors
- Mice
- Mice, Knockout
- Phosphorylation
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/physiology
- Signal Transduction/immunology
- Trans-Activators/genetics
- Trans-Activators/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
- Tyrosine/metabolism
Collapse
Affiliation(s)
- L A Garrett-Sinha
- Howard Hughes Medical Institute, University of Chicago, Illinois 60637, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Lloberas J, Soler C, Celada A. The key role of PU.1/SPI-1 in B cells, myeloid cells and macrophages. IMMUNOLOGY TODAY 1999; 20:184-9. [PMID: 10203717 DOI: 10.1016/s0167-5699(99)01442-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- J Lloberas
- Dept de Fisiologia (Biologia del macrofag), Facultat de Biologia, and Fundació August Pi i Sunyer, Campus Bellvitge, Universitat de Barcelona, Spain
| | | | | |
Collapse
|
47
|
Behre G, Whitmarsh AJ, Coghlan MP, Hoang T, Carpenter CL, Zhang DE, Davis RJ, Tenen DG. c-Jun is a JNK-independent coactivator of the PU.1 transcription factor. J Biol Chem 1999; 274:4939-46. [PMID: 9988737 DOI: 10.1074/jbc.274.8.4939] [Citation(s) in RCA: 152] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ETS domain transcription factor PU.1 is necessary for the development of monocytes and regulates, in particular, the expression of the monocyte-specific macrophage colony-stimulating factor (M-CSF) receptor, which is critical for monocytic cell survival, proliferation, and differentiation. The bZIP transcription factor c-Jun, which is part of the AP-1 transcription factor complex, is also important for monocytic differentiation, but the monocyte-specific M-CSF receptor promoter has no AP-1 consensus binding sites. We asked the question of whether c-Jun could promote the induction of the M-CSF receptor by collaborating with PU.1. We demonstrate that c-Jun enhances the ability of PU.1 to transactivate the M-CSF receptor promoter as well as a minimal thymidine kinase promoter containing only PU.1 DNA binding sites. c-Jun does not directly bind to the M-CSF receptor promoter but associates via its basic domain with the ETS domain of PU.1. Consistent with our observation that AP-1 binding does not contribute to c-Jun coactivation is the observation that the activation of PU.1 by c-Jun is blocked by overexpression of c-Fos. Phosphorylation of c-Jun by c-Jun NH2-terminal kinase on Ser-63 and -73 does not alter the ability of c-Jun to enhance PU.1 transactivation. Activated Ras enhances the transcriptional activity of PU.1 by up-regulating c-Jun expression without changing the phosphorylation pattern of PU.1. The activation of PU.1 by Ras is blocked by a mutant c-Jun protein lacking the basic domain. The expression of this mutant form of c-Jun also completely blocks 12-O-tetradecanoylphorbol-13-acetate-induced M-CSF receptor promoter activity during monocytic differentiation. We propose therefore that c-Jun acts as a c-Jun NH2-terminal kinase-independent coactivator of PU.1, resulting in M-CSF receptor expression and development of the monocytic lineage.
Collapse
Affiliation(s)
- G Behre
- Division of Hematology/Oncology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Brass AL, Zhu AQ, Singh H. Assembly requirements of PU.1-Pip (IRF-4) activator complexes: inhibiting function in vivo using fused dimers. EMBO J 1999; 18:977-91. [PMID: 10022840 PMCID: PMC1171190 DOI: 10.1093/emboj/18.4.977] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gene expression in higher eukaryotes appears to be regulated by specific combinations of transcription factors binding to regulatory sequences. The Ets factor PU.1 and the IRF protein Pip (IRF-4) represent a pair of interacting transcription factors implicated in regulating B cell-specific gene expression. Pip is recruited to its binding site on DNA by phosphorylated PU.1. PU.1-Pip interaction is shown to be template directed and involves two distinct protein-protein interaction surfaces: (i) the ets and IRF DNA-binding domains; and (ii) the phosphorylated PEST region of PU.1 and a lysine-requiring putative alpha-helix in Pip. Thus, a coordinated set of protein-protein and protein-DNA contacts are essential for PU.1-Pip ternary complex assembly. To analyze the function of these factors in vivo, we engineered chimeric repressors containing the ets and IRF DNA-binding domains connected by a flexible POU domain linker. When stably expressed, the wild-type fused dimer strongly repressed the expression of a rearranged immunoglobulin lambda gene, thereby establishing the functional importance of PU.1-Pip complexes in B cell gene expression. Comparative analysis of the wild-type dimer with a series of mutant dimers distinguished a gene regulated by PU.1 and Pip from one regulated by PU.1 alone. This strategy should prove generally useful in analyzing the function of interacting transcription factors in vivo, and for identifying novel genes regulated by such complexes.
Collapse
Affiliation(s)
- A L Brass
- Howard Hughes Medical Institute, The University of Chicago, 5841 S. Maryland Avenue, MC1028, Chicago IL 60637, USA
| | | | | |
Collapse
|
49
|
Ying H, Healy JI, Goodnow CC, Parnes JR. Regulation of Mouse CD72 Gene Expression During B Lymphocyte Development. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.9.4760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
CD72 is a 45-kDa transmembrane glycoprotein that is predominantly expressed on cells of the B lineage except plasma cells. Previously, we identified the 255-bp minimal mouse CD72 promoter capable of tissue-specific and developmental stage-specific expression. DNase I footprinting analysis of the 255-bp CD72 promoter revealed three protected elements, footprint (FP) I, FP II, and FP III. FP II, which extends from nucleotide −189 to −169 of the mouse CD72 promoter, exhibited both tissue-specific and developmental stage-specific activity that was reflective of the activity of the CD72 gene in vivo. In this report, we show that FP II is specifically recognized by the transcription factor B cell-specific activator protein (BSAP). Mutations eliminating the binding of BSAP in reporter constructs also eliminated the increase of reporter activity in B cells. In addition, cotransfections with reporter constructs plus different amounts of expression plasmids for BSAP showed that CD72 promoter activity was up-regulated by BSAP in plasmacytoma cells and T cells in a dose-dependent manner. Moreover, the expression level of CD72 decreased 10-fold on normal plasma cells. Compared with the presence of BSAP binding in mature B cells, the binding of BSAP was undetectable in those plasma cells. This study strongly suggests that BSAP-FP II interaction plays a critical role in determining the cell-type specificity of the CD72 promoter. The absence of positive factors such as BSAP accounts for at least part of the loss of mouse CD72 expression in plasma cells and thus might be common for the down-regulation of many molecules at the plasma cell stage.
Collapse
Affiliation(s)
- Han Ying
- *Division of Immunology and Rheumatology, Department of Medicine, and
| | - James I. Healy
- †Howard Hughes Medical Institute, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Christopher C. Goodnow
- †Howard Hughes Medical Institute, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - Jane R. Parnes
- *Division of Immunology and Rheumatology, Department of Medicine, and
| |
Collapse
|
50
|
Rao S, Karray S, Gackstetter ER, Koshland ME. Myocyte enhancer factor-related B-MEF2 is developmentally expressed in B cells and regulates the immunoglobulin J chain promoter. J Biol Chem 1998; 273:26123-9. [PMID: 9748293 DOI: 10.1074/jbc.273.40.26123] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Immunoglobulin J chain gene expression is induced by the delivery of a lymphokine signal to antigen-activated B cells in a primary immune response. A major interleukin 2 (IL-2)-responsive region that contains two adjacent control elements (JA and JB) exists within the J chain promoter. Transcription factor PU.1 positively regulates J chain gene expression by binding to one of the control elements (JB) in the J chain promoter. In the present study we have determined that a myocyte enhancer factor 2 (MEF2)-related nuclear factor, named B-MEF2, positively regulates the J chain gene promoter activity via the second control element (JA). An in vitro translated MEF2 family member, MEF2C, was found to bind the JA site with identical properties as endogenously expressed B-MEF2 in B cell lines. Moreover, in vivo experiments showed that a dominant negative mutant of MEF2C blocked B-MEF2 regulation of the J chain promoter. Consistent with its role as positive regulator of J chain gene expression, B-MEF2 levels were enhanced in highly differentiated B cells. In addition, induction of an IL-2-responsive presecretor cell line BCL1 with IL-2 or IL-5 (which up-regulates J chain gene expression) resulted in an increased expression of B-MEF2. We conclude that a MEF2-related transcriptional factor, B-MEF2, acts as a stage-specific positive regulator of J chain gene expression in the B cell lineage.
Collapse
Affiliation(s)
- S Rao
- Division of Immunology, Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA.
| | | | | | | |
Collapse
|