1
|
Mirzadeh Azad F, Struys EA, Wingert V, Hannibal L, Mills K, Jansen JH, Longley DB, Stunnenberg HG, Atlasi Y. Spic regulates one-carbon metabolism and histone methylation in ground-state pluripotency. SCIENCE ADVANCES 2023; 9:eadg7997. [PMID: 37595034 PMCID: PMC11801372 DOI: 10.1126/sciadv.adg7997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Understanding mechanisms of epigenetic regulation in embryonic stem cells (ESCs) is of fundamental importance for stem cell and developmental biology. Here, we identify Spic, a member of the ETS family of transcription factors (TFs), as a marker of ground state pluripotency. We show that Spic is rapidly induced in ground state ESCs and in response to extracellular signal-regulated kinase (ERK) inhibition. We find that SPIC binds to enhancer elements and stabilizes NANOG binding to chromatin, particularly at genes involved in choline/one-carbon (1C) metabolism such as Bhmt, Bhmt2, and Dmgdh. Gain-of-function and loss-of-function experiments revealed that Spic controls 1C metabolism and the flux of S-adenosyl methionine to S-adenosyl-L-homocysteine (SAM-to-SAH), thereby, modulating the levels of H3R17me2 and H3K4me3 histone marks in ESCs. Our findings highlight betaine-dependent 1C metabolism as a hallmark of ground state pluripotency primarily activated by SPIC. These findings underscore the role of uncharacterized auxiliary TFs in linking cellular metabolism to epigenetic regulation in ESCs.
Collapse
Affiliation(s)
- Fatemeh Mirzadeh Azad
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| | - Eduard A. Struys
- Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Victoria Wingert
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Luciana Hannibal
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ken Mills
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| | - Joop H. Jansen
- Department of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Daniel B. Longley
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| | - Hendrik G. Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, Netherlands
- Princess Maxima Centre for Pediatric Oncology, Utrecht, Netherlands
| | - Yaser Atlasi
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
2
|
Raczkowski HL, DeKoter RP. Lineage-instructive functions of the E26-transformation-specific-family transcription factor Spi-C in immune cell development and disease. WIREs Mech Dis 2021; 13:e1519. [PMID: 34730294 DOI: 10.1002/wsbm.1519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 11/10/2022]
Abstract
Cell fate decisions during hematopoiesis are the consequence of a complex mixture of inputs from cell-intrinsic and cell-extrinsic factors. In rare cases, expression of a single transcription factor, or a few key factors, may be sufficient to dictate lineage differentiation in a precursor cell. The E26-transformation-specific-family transcription factor Spi-C has emerged as an example of a lineage-instructive factor involved in the generation of mature, specialized subsets of both myeloid and lymphoid cells. Spi-C can instruct differentiation of splenic precursors into red pulp macrophages responsible for phagocytosing senescent red blood cells. In the B cell compartment, Spi-C acts as a key regulator of cell fate decisions at the pro-B to pre-B cell stage and for plasma cell differentiation. Spi-C regulates key genes including Nfkb1, Bach2, Syk, and Blnk to regulate cell cycle entry and B cell differentiation. Here, we review the biology of the lineage-instructive transcription factor Spi-C and its contribution to mechanisms of disease in macrophages and B cells. This article is categorized under: Cancer > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology Infectious Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Hannah L Raczkowski
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario, Canada
| | - Rodney P DeKoter
- Department of Microbiology & Immunology and the Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario, Canada
| |
Collapse
|
3
|
Niidome K, Taniguchi R, Yamazaki T, Tsuji M, Itoh K, Ishihara Y. FosL1 Is a Novel Target of Levetiracetam for Suppressing the Microglial Inflammatory Reaction. Int J Mol Sci 2021; 22:ijms222010962. [PMID: 34681621 PMCID: PMC8537483 DOI: 10.3390/ijms222010962] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Abstract
We previously showed that the antiepileptic drug levetiracetam (LEV) inhibits microglial activation, but the mechanism remains unclear. The purpose of this study was to identify the target of LEV in microglial activity suppression. The mouse microglial BV-2 cell line, cultured in a ramified form, was pretreated with LEV and then treated with lipopolysaccharide (LPS). A comprehensive analysis of LEV targets was performed by cap analysis gene expression sequencing using BV-2 cells, indicating the transcription factors BATF, Nrf-2, FosL1 (Fra1), MAFF, and Spic as candidates. LPS increased AP-1 and Spic transcriptional activity, and LEV only suppressed AP-1 activity. FosL1, MAFF, and Spic mRNA levels were increased by LPS, and LEV only attenuated FosL1 mRNA expression, suggesting FosL1 as an LEV target. FosL1 protein levels were increased by LPS treatment and decreased by LEV pretreatment, similar to FosL1 mRNA levels. The FosL1 siRNA clearly suppressed the expression of TNFα and IL-1β. Pilocarpine-induced status epilepticus increased hippocampus FosL1 expression, along with inflammation. LEV treatment significantly suppressed FosL1 expression. Together, LEV reduces FosL1 expression and AP-1 activity in activated microglia, thereby suppressing neuroinflammation. LEV might be a candidate for the treatment of several neurological diseases involving microglial activation.
Collapse
Affiliation(s)
- Kouji Niidome
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (K.N.); (R.T.)
| | - Ruri Taniguchi
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (K.N.); (R.T.)
| | - Takeshi Yamazaki
- Program of Life and Environmental Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan;
| | - Mayumi Tsuji
- Department of Environmental Health, University of Occupational and Environmental Health, Fukuoka 807-8555, Japan;
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Kagawa Bunri University, Sanuki 769-219, Japan;
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan; (K.N.); (R.T.)
- Correspondence:
| |
Collapse
|
4
|
RAG-Mediated DNA Breaks Attenuate PU.1 Activity in Early B Cells through Activation of a SPIC-BCLAF1 Complex. Cell Rep 2020; 29:829-843.e5. [PMID: 31644907 PMCID: PMC6870970 DOI: 10.1016/j.celrep.2019.09.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/10/2019] [Accepted: 09/09/2019] [Indexed: 11/22/2022] Open
Abstract
Early B cell development is regulated by stage-specific transcription
factors. PU.1, an ETS-family transcription factor, is essential for coordination
of early B cell maturation and immunoglobulin gene (Ig)
rearrangement. Here we show that RAG DNA double-strand breaks (DSBs) generated
during Ig light chain gene (Igl) rearrangement
in pre-B cells induce global changes in PU.1 chromatin binding. RAG DSBs
activate a SPIC/BCLAF1 transcription factor complex that displaces PU.1
throughout the genome and regulates broad transcriptional changes. SPIC recruits
BCLAF1 to gene-regulatory elements that control expression of key B cell
developmental genes. The SPIC/BCLAF1 complex suppresses expression of the SYK
tyrosine kinase and enforces the transition from large to small pre-B cells.
These studies reveal that RAG DSBs direct genome-wide changes in ETS
transcription factor activity to promote early B cell development. ETS-family transcription factors are key regulators of early B cell
development. Soodgupta et al. show that RAG-induced DNA breaks generated during
antigen receptor gene recombination activate a SPIC/BCLAF1 transcription factor
complex that counters PU.1 activity and regulates gene expression changes to
promote transition from large to small pre-B cells.
Collapse
|
5
|
Laramée AS, Raczkowski H, Shao P, Batista C, Shukla D, Xu L, Haeryfar SMM, Tesfagiorgis Y, Kerfoot S, DeKoter R. Opposing Roles for the Related ETS-Family Transcription Factors Spi-B and Spi-C in Regulating B Cell Differentiation and Function. Front Immunol 2020; 11:841. [PMID: 32457757 PMCID: PMC7225353 DOI: 10.3389/fimmu.2020.00841] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Generation of specific antibodies during an immune response to infection or vaccination depends on the ability to rapidly and accurately select clones of antibody-secreting B lymphocytes for expansion. Antigen-specific B cell clones undergo the cell fate decision to differentiate into antibody-secreting plasma cells, memory B cells, or germinal center B cells. The E26-transformation-specific (ETS) transcription factors Spi-B and Spi-C are important regulators of B cell development and function. Spi-B is expressed throughout B cell development and is downregulated upon plasma cell differentiation. Spi-C is highly related to Spi-B and has similar DNA-binding specificity. Heterozygosity for Spic rescues B cell development and B cell proliferation defects observed in Spi-B knockout mice. In this study, we show that heterozygosity for Spic rescued defective IgG1 secondary antibody responses in Spib–/– mice. Plasma cell differentiation was accelerated in Spib–/– B cells. Gene expression, ChIP-seq, and reporter gene analysis showed that Spi-B and Spi-C differentially regulated Bach2, encoding a key regulator of plasma cell and memory B cell differentiation. These results suggest that Spi-B and Spi-C oppose the function of one another to regulate B cell differentiation and function.
Collapse
Affiliation(s)
- Anne-Sophie Laramée
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Hannah Raczkowski
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Peng Shao
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Carolina Batista
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - Devanshi Shukla
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Li Xu
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON, Canada
| | - Yodit Tesfagiorgis
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Steven Kerfoot
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Rodney DeKoter
- Department of Microbiology and Immunology, Center for Human Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.,Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, ON, Canada
| |
Collapse
|
6
|
Spi-C positively regulates RANKL-mediated osteoclast differentiation and function. Exp Mol Med 2020; 52:691-701. [PMID: 32341419 PMCID: PMC7210314 DOI: 10.1038/s12276-020-0427-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022] Open
Abstract
Spi-C is an SPI-group erythroblast transformation-specific domain transcription factor expressed during B-cell development. Here, we report that Spi-C is a novel receptor activator of nuclear factor-κB ligand (RANKL)-inducible protein that positively regulates RANKL-mediated osteoclast differentiation and function. Knockdown of Spi-C decreased the expression of RANKL-induced nuclear factor of activated T-cells, cytoplasmic 1, receptor activator of nuclear factor-κB (RANK), and tartrate-resistant acid phosphatase (TRAP), resulting in a marked decrease in the number of TRAP-positive multinucleated cells. Spi-C-transduced bone marrow-derived monocytes/macrophages (BMMs) displayed a significant increase in osteoclast formation in the presence of RANKL. In addition, Spi-C-depleted cells failed to show actin ring formation or bone resorption owing to a marked reduction in the expression of RANKL-mediated dendritic cell-specific transmembrane protein and the d2 isoform of vacuolar (H+) ATPase V0 domain, which are known osteoclast fusion-related genes. Interestingly, RANKL stimulation induced the translocation of Spi-C from the cytoplasm into the nucleus during osteoclastogenesis, which was specifically blocked by inhibitors of p38 mitogen-activated protein kinase (MAPK) or PI3 kinase. Moreover, Spi-C depletion prevented RANKL-induced MAPK activation and the degradation of inhibitor of κB-α (IκBα) in BMMs. Collectively, these results suggest that Spi-C is a novel positive regulator that promotes both osteoclast differentiation and function. A gene-controlling protein called Spi-C promotes the development of bone-processing cells called osteoclasts; details of the molecular mechanisms involved will aid understanding of Spi-C’s role in bone health and disease. Osteoclasts degrade bone during the normal process of bone remodeling, balanced by the activity of osteoblast cells that form new bone. Excessive osteoclast activity can cause the bone loss associated with various bone diseases including early-onset osteoporosis. Researchers in South Korea led by Soo Young Lee at Ewha Womans University and Na Kyung Lee at Soonchunhyang University, Asan, found that Spi-C promotes osteoclast development by activating genes that code for key proteins of a signaling pathway known to be crucial for bone health. Drugs that interfere with Spi-C activity may therefore offer a new approach for treating bone disease.
Collapse
|
7
|
Abstract
DNA damage occurs on exposure to genotoxic agents and during physiological DNA transactions. DNA double-strand breaks (DSBs) are particularly dangerous lesions that activate DNA damage response (DDR) kinases, leading to initiation of a canonical DDR (cDDR). This response includes activation of cell cycle checkpoints and engagement of pathways that repair the DNA DSBs to maintain genomic integrity. In adaptive immune cells, programmed DNA DSBs are generated at precise genomic locations during the assembly and diversification of lymphocyte antigen receptor genes. In innate immune cells, the production of genotoxic agents, such as reactive nitrogen molecules, in response to pathogens can also cause genomic DNA DSBs. These DSBs in adaptive and innate immune cells activate the cDDR. However, recent studies have demonstrated that they also activate non-canonical DDRs (ncDDRs) that regulate cell type-specific processes that are important for innate and adaptive immune responses. Here, we review these ncDDRs and discuss how they integrate with other signals during immune system development and function.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Barry P Sleckman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Heme ameliorates dextran sodium sulfate-induced colitis through providing intestinal macrophages with noninflammatory profiles. Proc Natl Acad Sci U S A 2018; 115:8418-8423. [PMID: 30061415 PMCID: PMC6099887 DOI: 10.1073/pnas.1808426115] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Following hemorrhage in damaged tissues, hemoglobin induces macrophages (Mϕs) possessing ability to protect against tissue inflammation. Hemorrhage-appearing mucosa is observed in patients with inflammatory bowel disease. However, heme-mediated modulation of intestinal Mϕ activity remains poorly understood. Here, we provide evidence that Spi-C induced by heme is a key molecule for providing noninflammatory gene expression patterns of intestinal CX3CR1high Mϕs. We found that the Spic deficiency in intestinal Mϕs resulted in increased sensitivity to dextran sodium sulfate-induced colitis. Heme-mediated Spi-C inhibited a subset of LPS-induced genes such as Il6 and Il1a by intestinal CX3CR1high Mϕs through inhibition of IRF5-NF-κB p65 complex formation. These results reveal a mechanism modulating the noninflammatory phenotype of intestinal Mϕs and may help identify targets for therapy of intestinal inflammation. The local environment is crucial for shaping the identities of tissue-resident macrophages (Mϕs). When hemorrhage occurs in damaged tissues, hemoglobin induces differentiation of anti-inflammatory Mϕs with reparative function. Mucosal bleeding is one of the pathological features of inflammatory bowel diseases. However, the heme-mediated mechanism modulating activation of intestinal innate immune cells remains poorly understood. Here, we show that heme regulates gut homeostasis through induction of Spi-C in intestinal CX3CR1high Mϕs. Intestinal CX3CR1high Mϕs highly expressed Spi-C in a heme-dependent manner, and myeloid lineage-specific Spic-deficient (Lyz2-cre; Spicflox/flox) mice showed severe intestinal inflammation with an increased number of Th17 cells during dextran sodium sulfate-induced colitis. Spi-C down-regulated the expression of a subset of Toll-like receptor (TLR)-inducible genes in intestinal CX3CR1high Mϕs to prevent colitis. LPS-induced production of IL-6 and IL-1α, but not IL-10 and TNF-α, by large intestinal Mϕs from Lyz2-cre; Spicflox/flox mice was markedly enhanced. The interaction of Spi-C with IRF5 was linked to disruption of the IRF5-NF-κB p65 complex formation, thereby abrogating recruitment of IRF5 and NF-κB p65 to the Il6 and Il1a promoters. Collectively, these results demonstrate that heme-mediated Spi-C is a key molecule for the noninflammatory signature of intestinal Mϕs by suppressing the induction of a subset of TLR-inducible genes through binding to IRF5.
Collapse
|
9
|
Alam MZ, Devalaraja S, Haldar M. The Heme Connection: Linking Erythrocytes and Macrophage Biology. Front Immunol 2017; 8:33. [PMID: 28167947 PMCID: PMC5256077 DOI: 10.3389/fimmu.2017.00033] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022] Open
Abstract
Erythroid function and development is intimately linked to macrophages. The primary function of erythrocytes is oxygen delivery, which is mediated by iron-containing hemoglobin. The major source of this iron is a recycling pathway where macrophages scavenge old and damaged erythrocytes to release iron contained within the heme moiety. Macrophages also promote erythropoiesis by providing a supportive niche in the bone marrow as an integral component of “erythorblastic islands.” Importantly, inflammation leads to alterations in iron handling by macrophages with significant impact on iron homeostasis and erythropoiesis. The importance of macrophages in erythropoiesis and iron homeostasis is well established and has been extensively reviewed. However, this developmental relationship is not one way, and erythrocytes can also regulate macrophage development and function. Erythrocyte-derived heme can induce the development of iron-recycling macrophages from monocytes, engage pattern recognition receptors to activate macrophages, and act as ligand for specific nuclear receptors to modulate macrophage function. Here, we discuss the role of heme as a signaling molecule impacting macrophage homeostasis. We will review these actions of heme within the framework of our current understanding of the role of micro-environmental factors in macrophage development and function.
Collapse
Affiliation(s)
- Md Zahidul Alam
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| | - Samir Devalaraja
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| | - Malay Haldar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
10
|
Bednarski JJ, Pandey R, Schulte E, White LS, Chen BR, Sandoval GJ, Kohyama M, Haldar M, Nickless A, Trott A, Cheng G, Murphy KM, Bassing CH, Payton JE, Sleckman BP. RAG-mediated DNA double-strand breaks activate a cell type-specific checkpoint to inhibit pre-B cell receptor signals. J Exp Med 2016; 213:209-23. [PMID: 26834154 PMCID: PMC4749927 DOI: 10.1084/jem.20151048] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/03/2015] [Indexed: 01/17/2023] Open
Abstract
DNA double-strand breaks (DSBs) activate a canonical DNA damage response, including highly conserved cell cycle checkpoint pathways that prevent cells with DSBs from progressing through the cell cycle. In developing B cells, pre-B cell receptor (pre-BCR) signals initiate immunoglobulin light (Igl) chain gene assembly, leading to RAG-mediated DNA DSBs. The pre-BCR also promotes cell cycle entry, which could cause aberrant DSB repair and genome instability in pre-B cells. Here, we show that RAG DSBs inhibit pre-BCR signals through the ATM- and NF-κB2-dependent induction of SPIC, a hematopoietic-specific transcriptional repressor. SPIC inhibits expression of the SYK tyrosine kinase and BLNK adaptor, resulting in suppression of pre-BCR signaling. This regulatory circuit prevents the pre-BCR from inducing additional Igl chain gene rearrangements and driving pre-B cells with RAG DSBs into cycle. We propose that pre-B cells toggle between pre-BCR signals and a RAG DSB-dependent checkpoint to maintain genome stability while iteratively assembling Igl chain genes.
Collapse
Affiliation(s)
- Jeffrey J Bednarski
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Ruchi Pandey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Emily Schulte
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Lynn S White
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Bo-Ruei Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gabriel J Sandoval
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Masako Kohyama
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Malay Haldar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew Nickless
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Amanda Trott
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Barry P Sleckman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
11
|
Kierdorf K, Prinz M, Geissmann F, Gomez Perdiguero E. Development and function of tissue resident macrophages in mice. Semin Immunol 2015; 27:369-78. [PMID: 27036090 PMCID: PMC4948121 DOI: 10.1016/j.smim.2016.03.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 12/24/2022]
Abstract
Macrophages are important for tissue development, homeostasis as well as immune response upon injury or infection. For a long time they were only seen as one uniform group of phagocytes with a common origin and similar functions. However, this view has been challenged in the last decade and revealed a complex diversity of tissue resident macrophages. Here, we want to present the current view on macrophage development and tissue specification and we will discuss differences as well as common patterns between heterogeneous macrophage subpopulations.
Collapse
Affiliation(s)
- Katrin Kierdorf
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King's College London, London, UK
| | - Marco Prinz
- Institute of Neuropathology, University Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Frederic Geissmann
- Centre for Molecular and Cellular Biology of Inflammation (CMCBI), King's College London, London, UK; Immunology Program, Memorial Sloan Kettering Cancer Center, NY, NY, USA
| | - Elisa Gomez Perdiguero
- Macrophages and Endothelial Cells group, Department of Developmental and Stem Cell Biology, CNRS UMR 3738, Institut Pasteur, Paris, France.
| |
Collapse
|
12
|
Li SKH, Solomon LA, Fulkerson PC, DeKoter RP. Identification of a negative regulatory role for spi-C in the murine B cell lineage. THE JOURNAL OF IMMUNOLOGY 2015; 194:3798-807. [PMID: 25769919 DOI: 10.4049/jimmunol.1402432] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022]
Abstract
Spi-C is an E26 transformation-specific family transcription factor that is highly related to PU.1 and Spi-B. Spi-C is expressed in developing B cells, but its function in B cell development and function is not well characterized. To determine whether Spi-C functions as a negative regulator of Spi-B (encoded by Spib), mice were generated that were germline knockout for Spib and heterozygous for Spic (Spib(-/-)Spic(+/-)). Interestingly, loss of one Spic allele substantially rescued B cell frequencies and absolute numbers in Spib(-/-) mouse spleens. Spib(-/-)Spic(+/-) B cells had restored proliferation compared with Spib(-/-) B cells in response to anti-IgM or LPS stimulation. Investigation of a potential mechanism for the Spib(-/-)Spic(+/-) phenotype revealed that steady-state levels of Nfkb1, encoding p50, were elevated in Spib(-/-)Spic(+/-) B cells compared with Spib(-/-) B cells. Spi-B was shown to directly activate the Nfkb1 gene, whereas Spi-C was shown to repress this gene. These results indicate a novel role for Spi-C as a negative regulator of B cell development and function.
Collapse
Affiliation(s)
- Stephen K H Li
- Department of Microbiology and Immunology, Centre for Human Immunology, Schulich School of Medicine and Dentistry, Collaborative Graduate Program in Developmental Biology, Western University, London, Ontario N6A 5C1, Canada; Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario N6C 2V5, Canada; and
| | - Lauren A Solomon
- Department of Microbiology and Immunology, Centre for Human Immunology, Schulich School of Medicine and Dentistry, Collaborative Graduate Program in Developmental Biology, Western University, London, Ontario N6A 5C1, Canada; Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario N6C 2V5, Canada; and
| | - Patricia C Fulkerson
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Rodney P DeKoter
- Department of Microbiology and Immunology, Centre for Human Immunology, Schulich School of Medicine and Dentistry, Collaborative Graduate Program in Developmental Biology, Western University, London, Ontario N6A 5C1, Canada; Division of Genetics and Development, Children's Health Research Institute, Lawson Research Institute, London, Ontario N6C 2V5, Canada; and
| |
Collapse
|
13
|
de Almeida CR, Hendriks RW, Stadhouders R. Dynamic Control of Long-Range Genomic Interactions at the Immunoglobulin κ Light-Chain Locus. Adv Immunol 2015; 128:183-271. [DOI: 10.1016/bs.ai.2015.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Heme-mediated SPI-C induction promotes monocyte differentiation into iron-recycling macrophages. Cell 2014; 156:1223-1234. [PMID: 24630724 DOI: 10.1016/j.cell.2014.01.069] [Citation(s) in RCA: 322] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 11/29/2013] [Accepted: 01/23/2014] [Indexed: 02/06/2023]
Abstract
Splenic red pulp macrophages (RPM) degrade senescent erythrocytes and recycle heme-associated iron. The transcription factor SPI-C is selectively expressed by RPM and is required for their development, but the physiologic stimulus inducing Spic is unknown. Here, we report that Spic also regulated the development of F4/80(+)VCAM1(+) bone marrow macrophages (BMM) and that Spic expression in BMM and RPM development was induced by heme, a metabolite of erythrocyte degradation. Pathologic hemolysis induced loss of RPM and BMM due to excess heme but induced Spic in monocytes to generate new RPM and BMM. Spic expression in monocytes was constitutively inhibited by the transcriptional repressor BACH1. Heme induced proteasome-dependent BACH1 degradation and rapid Spic derepression. Furthermore, cysteine-proline dipeptide motifs in BACH1 that mediate heme-dependent degradation were necessary for Spic induction by heme. These findings are the first example of metabolite-driven differentiation of a tissue-resident macrophage subset and provide new insights into iron homeostasis.
Collapse
|
15
|
DeKoter RP, Geadah M, Khoosal S, Xu LS, Thillainadesan G, Torchia J, Chin SS, Garrett-Sinha LA. Regulation of Follicular B Cell Differentiation by the Related E26 Transformation-Specific Transcription Factors PU.1, Spi-B, and Spi-C. THE JOURNAL OF IMMUNOLOGY 2010; 185:7374-84. [DOI: 10.4049/jimmunol.1001413] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
16
|
Imipramine treatment and resiliency exhibit similar chromatin regulation in the mouse nucleus accumbens in depression models. J Neurosci 2009; 29:7820-32. [PMID: 19535594 DOI: 10.1523/jneurosci.0932-09.2009] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although it is a widely studied psychiatric syndrome, major depressive disorder remains a poorly understood illness, especially with regard to the disconnect between treatment initiation and the delayed onset of clinical improvement. We have recently validated chronic social defeat stress in mice as a model in which a depression-like phenotype is reversed by chronic, but not acute, antidepressant administration. Here, we use chromatin immunoprecipitation (ChIP)-chip assays--ChIP followed by genome wide promoter array analyses--to study the effects of chronic defeat stress on chromatin regulation in the mouse nucleus accumbens (NAc), a key brain reward region implicated in depression. Our results demonstrate that chronic defeat stress causes widespread and long-lasting changes in gene regulation, including alterations in repressive histone methylation and in phospho-CREB (cAMP response element-binding protein) binding, in the NAc. We then show similarities and differences in this regulation to that observed in another mouse model of depression, prolonged adult social isolation. In the social defeat model, we observed further that many of the stress-induced changes in gene expression are reversed by chronic imipramine treatment, and that resilient mice-those resistant to the deleterious effects of defeat stress-show patterns of chromatin regulation in the NAc that overlap dramatically with those seen with imipramine treatment. These findings provide new insight into the molecular basis of depression-like symptoms and the mechanisms by which antidepressants exert their delayed clinical efficacy. They also raise the novel idea that certain individuals resistant to stress may naturally mount antidepressant-like adaptations in response to chronic stress.
Collapse
|
17
|
Role for Spi-C in the development of red pulp macrophages and splenic iron homeostasis. Nature 2008; 457:318-21. [PMID: 19037245 PMCID: PMC2756102 DOI: 10.1038/nature07472] [Citation(s) in RCA: 343] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Accepted: 09/30/2008] [Indexed: 12/27/2022]
Abstract
Tissue macrophages comprise a heterogeneous group of cell types differing in location, surface markers and function1. Red pulp macrophages are a distinct splenic subset involved in removing senescent red blood cells2. Transcription factors such as PU.1 and C/EBPα play general roles in myelomonocytic development3,4, but the transcriptional basis for producing tissue macrophage subsets remains unknown. Here we show that Spi-C, a PU.1 related transcription factor, selectively controls the development of red pulp macrophages. Spi-C is highly expressed in red pulp macrophages, but not monocytes, dendritic cells or other tissue macrophages. Spi-C−/− mice exhibit a cell-autonomous defect in the development of red pulp macrophages that is corrected by retroviral Spi-C expression in bone marrow cells, but have normal monocyte and other macrophage subsets. Red pulp macrophages highly express genes involved in capturing circulating hemoglobin and iron regulation. Spi-C−/− mice show normal trapping of red blood cells in the spleen, but fail to phagocytose these red blood cells efficiently, and develop an iron overload localized selectively to splenic red pulp. Thus, Spi-C controls development of red pulp macrophages required for red blood cell recycling and iron homeostasis.
Collapse
|
18
|
Zhu X, Schweitzer BL, Romer EJ, Sulentic CEW, DeKoter RP. Transgenic expression of Spi-C impairs B-cell development and function by affecting genes associated with BCR signaling. Eur J Immunol 2008; 38:2587-99. [PMID: 18792411 DOI: 10.1002/eji.200838323] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Spi-C is an Ets family transcription factor closely related to PU.1 and Spi-B. Expression of Spi-C is developmentally regulated in the B-cell lineage, but its function remains unknown. To determine the function of Spi-C in B-cell development, we generated mice expressing a B-cell-specific Spi-C transgene under the control of the IgH intronic enhancer. Spi-C transgenic mice had 50% fewer B cells than wild-type littermates. Flow cytometric analyses showed that splenic transitional B cells and bone marrow pre-B or immature B cells from transgenic mice were dramatically reduced compared with those of wild type. Both nonspecific and Ag-specific serum IgM levels were significantly increased in transgenic mice, while serum IgG levels were significantly decreased compared with wild type. Spi-C transgenic B cells proliferated poorly after stimulation by anti-IgM or anti-CD40 in vitro, although they responded normally to LPS stimulation. Using real-time RT-PCR, we found that several BCR signaling-related mediators were downregulated at pre-B-cell and mature B-cell stages in transgenic mice, while an inhibitor of BCR signaling was upregulated. Taken together, these data indicate that ectopic expression of Spi-C can impair B-cell development and function by affecting genes associated with BCR signaling.
Collapse
Affiliation(s)
- Xiang Zhu
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0524, USA
| | | | | | | | | |
Collapse
|
19
|
Debnath I, Roundy KM, Dunn DM, Weiss RB, Weis JJ, Weis JH. Defining a transcriptional fingerprint of murine splenic B-cell development. Genes Immun 2008; 9:706-20. [PMID: 18784731 DOI: 10.1038/gene.2008.70] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
B-cell development occurs in a stepwise fashion that can be followed by the expression of B cell-specific surface markers. In this study, we wished to identify proteins that could contribute to the changes in expression of such markers. By using RNA from freshly isolated B220+ cells, we hoped to reduce the effect of artifacts that occur during the isolation and amplification steps necessary to use flow cytometry analysis-sorted subsets in microarray experiments. Analyses comparing expression patterns from B220+ 2-week bone marrow (pro-B, pre-B, immature B cells), 2-week spleen (predominantly transitional cells) and 8-week spleen (mainly mature B cells) yielded hundreds of genes. We also examined the B cell-activating factor (BAFF)-dependent effects on immature splenic B cells by comparing expression patterns in the spleen between 2-week A/J vs 2-week A/WySnJ mice, which lack functional BAFF receptor signaling. Genes that showed the expression differences between spleen and bone marrow samples were then analyzed through quantitative PCR on B-cell subsets isolated using two different sorting protocols. A comparison of the results from our study with the results from other analyses showed not only some overlap of preferentially expressed genes but also an expansion of other genes potentially involved in B-cell development.
Collapse
Affiliation(s)
- I Debnath
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | | | | | |
Collapse
|
20
|
Yan C, Lian X, Li Y, Dai Y, White A, Qin Y, Li H, Hume DA, Du H. Macrophage-specific expression of human lysosomal acid lipase corrects inflammation and pathogenic phenotypes in lal-/- mice. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 169:916-26. [PMID: 16936266 PMCID: PMC1698822 DOI: 10.2353/ajpath.2006.051327] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lysosomal acid lipase (LAL) hydrolyzes cholesteryl esters and triglycerides to generate free fatty acids and cholesterol in the cell. The downstream metabolites of these compounds serve as hormonal ligands for nuclear receptors and transcription factors. Genetic ablation of the lal gene in the mouse caused malformation of macrophages and inflammation-triggered multiple pathogenic phenotypes in multiple organs. To assess the relationship between macrophages and lal-/- pathogenic phenotypes, a macrophage-specific doxycycline-inducible transgenic system was generated to induce human LAL (hLAL) expression in the lal-/- genetic background under control of the 7.2-kb c-fms promoter/intron2 regulatory sequence. Doxycycline-induced hLAL expression in macrophages significantly ameliorated aberrant gene expression, inflammatory cell (neutrophil) influx, and pathogenesis in multiple organs. These studies strongly support that neutral lipid metabolism in macrophages contributes to organ inflammation and pathogenesis.
Collapse
Affiliation(s)
- Cong Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, and Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
Pritsker M, Ford NR, Jenq HT, Lemischka IR. Genomewide gain-of-function genetic screen identifies functionally active genes in mouse embryonic stem cells. Proc Natl Acad Sci U S A 2006; 103:6946-51. [PMID: 16621925 PMCID: PMC1458999 DOI: 10.1073/pnas.0509861103] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Indexed: 11/18/2022] Open
Abstract
Embryonic stem (ES) cells hold great promise for the future of medicine. To elucidate the molecular mechanisms that control ES cell self-renewal and differentiation, a comprehensive knowledge of the molecules involved in these processes is required. Here we describe an effective approach for genomewide identification of functionally active genes in ES cells. This approach combines genetic screens based on cDNA libraries with microarray detection methods to permit high-throughput functional analyses. We implement this strategy to identify genes whose overexpression can maintain phenotypic properties of undifferentiated mouse ES cells under differentiation-inducing conditions, specifically in the absence of leukemia inhibitory factor. The identified genes encode a variety of regulatory proteins whose function in ES cells was previously unknown. Moreover, our approach is capable of detecting genes whose overexpression promote differentiation or cell death. Overall, our studies establish a methodology for highly sensitive identification of genes that confer particular phenotypes on ES cells.
Collapse
Affiliation(s)
- Moshe Pritsker
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Nicole R. Ford
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Harry T. Jenq
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | - Ihor R. Lemischka
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
23
|
Carlsson R, Thorell K, Liberg D, Leanderson T. SPI-C and STAT6 can cooperate to stimulate IgE germline transcription. Biochem Biophys Res Commun 2006; 344:1155-60. [PMID: 16647686 DOI: 10.1016/j.bbrc.2006.04.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Accepted: 04/04/2006] [Indexed: 12/18/2022]
Abstract
SPI-C is a novel ETS protein that is expressed in B lymphocytes. No target gene for SPI-C has so far been defined. We have performed a yeast two-hybrid screen using SPI-C as bait in order to further analyze the functional role of this orphan transcription factor. We found that SPI-C interacted specifically with the C-terminus of STAT6 in yeast. By co-immunoprecipitation in transfected COS7 cells the physical interaction between SPI-C and STAT6 was confirmed. Furthermore, this protein-protein interaction is functional since we could demonstrate that SPI-C and STAT6 stimulated IL4 induced Iepsilon transcription synergistically but only when both proteins bound to DNA. Thus, a protein interaction between SPI-C and STAT6 is the basis for a novel mechanism for regulation of IL4 induced gene expression.
Collapse
Affiliation(s)
- Robert Carlsson
- Immunology Group, Lund University, BMC I:13, 22184 Lund, Sweden
| | | | | | | |
Collapse
|
24
|
Hagman J, Lukin K. Transcription factors drive B cell development. Curr Opin Immunol 2006; 18:127-34. [PMID: 16464566 DOI: 10.1016/j.coi.2006.01.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 01/25/2006] [Indexed: 11/25/2022]
Abstract
Transcription factors including PU.1, E2A and early B cell factor (EBF) are essential for the earliest stages of B lymphocyte development. Recent advances suggest that, although PU.1 initiates events leading to B lymphopoiesis, it might be dispensable at later stages of development. E2A proteins are also crucial for B cell lineage determination, as shown by the pluripotency of E2A-deficient progenitors. Both PU.1 and E2A are required for expression of EBF. EBF activates the early program of genes unique to B cells, including the lineage commitment factor Pax5. EBF also facilitates the function of Pax5 by mediating epigenetic changes necessary for the function of Pax5 at gene targets. Together, these proteins function in a hierarchy of factors that orchestrates B cell development.
Collapse
Affiliation(s)
- James Hagman
- Integrated Department of Immunology, National Jewish Medical and Research Center, 1400 Jackson Street, K516B, Denver, CO 80206, USA.
| | | |
Collapse
|
25
|
Abstract
Transcriptional regulation of T-cell development involves successive interactions between complexes of transcriptional regulators and their binding sites within the regulatory regions of each gene. The regulatory modules that control expression of T-lineage genes frequently include binding sites for a core set of regulators that set the T-cell-specific background for signal-dependent control, including GATA-3, Notch/CSL, c-myb, TCF-1, Ikaros, HEB/E2A, Ets, and Runx factors. Additional regulators in early thymocytes include PU.1, Id-2, SCL, Spi-B, Erg, Gfi-1, and Gli. Many of these factors are involved in simultaneous regulation of non-T-lineage genes, T-lineage genes, and genes involved in cell cycle control, apoptosis, or survival. Potential and known interactions between early thymic transcription factors such as GATA-3, SCL, PU.1, Erg, and Spi-B are explored. Regulatory modules involved in the expression of several critical T-lineage genes are described, and models are presented for shifting occupancy of the DNA-binding sites in the regulatory modules of pre-Talpha, T-cell receptor beta (TCRbeta), recombinase activating genes 1 and 2 (Rag-1/2), and CD4 during T-cell development. Finally, evidence is presented that c-kit, Erg, Hes-1, and HEBAlt are expressed differently in Rag-2(-/-) thymocytes versus normal early thymocytes, which provide insight into potential regulatory interactions that occur during normal T-cell development.
Collapse
Affiliation(s)
- Michele K Anderson
- Sunnybrook and Women's College Health Sciences Center, Division of Molecular and Cell Biology, University of Toronto, Department of Immunology, Toronto, ON, Canada.
| |
Collapse
|
26
|
Lian X, Yan C, Qin Y, Knox L, Li T, Du H. Neutral lipids and peroxisome proliferator-activated receptor-{gamma} control pulmonary gene expression and inflammation-triggered pathogenesis in lysosomal acid lipase knockout mice. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:813-21. [PMID: 16127159 PMCID: PMC1698726 DOI: 10.1016/s0002-9440(10)62053-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The functional roles of neutral lipids in the lung are poorly understood. However, blocking cholesteryl ester and triglyceride metabolism in lysosomal acid lipase gene knockout mice (lal-/-) results in severe pathogenic phenotypes in the lung, including massive neutrophil infiltration, foamy macrophage accumulation, unwanted cell growth, and emphysema. To elucidate the mechanism underlining these pathologies, we performed Affymetrix GeneChip microarray analysis of 1-, 3-, and 6-month-old mice and identified aberrant gene expression that progressed with age. Among changed genes, matrix metalloproteinase (MMP)-12, apoptosis inhibitor 6 (Api-6), erythroblast transformation-specific domain (Ets) transcription factor family member Spi-C, and oncogene MafB were increased 100-, 70-, 40-, and 10-fold, respectively, in lal-/- lungs versus the wild-type lungs. The pathogenic increases of these molecules occurred primarily in alveolar type II epithelial cells. Transcriptional activities of the MMP-12 and Api-6 promoters were stimulated by Spi-C or MafB in respiratory epithelial cells. Treatment with 9-hydroxyoctadecanoic acids and ciglitazone significantly rescued lal-/- pulmonary inflammation and aberrant gene expression. In addition, both compounds as well as peroxisome proliferator-activated receptor gamma inhibited MMP-12 and Api-6 promoter activities. These data suggest that inflammation-triggered cell growth and emphysema during lysosomal acid lipase deficiency are partially caused by peroxisome proliferator-activated receptor-gamma inactivation.
Collapse
Affiliation(s)
- Xuemei Lian
- Division of Pulmonary Biology, or Hong Du, Ph.D., Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | |
Collapse
|
27
|
Anderson MK, Pant R, Miracle AL, Sun X, Luer CA, Walsh CJ, Telfer JC, Litman GW, Rothenberg EV. Evolutionary origins of lymphocytes: ensembles of T cell and B cell transcriptional regulators in a cartilaginous fish. THE JOURNAL OF IMMUNOLOGY 2004; 172:5851-60. [PMID: 15128764 DOI: 10.4049/jimmunol.172.10.5851] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The evolutionary origins of lymphocytes can be traced by phylogenetic comparisons of key features. Homologs of rearranging TCR and Ig (B cell receptor) genes are present in jawed vertebrates, but have not been identified in other animal groups. In contrast, most of the transcription factors that are essential for the development of mammalian T and B lymphocytes belong to multigene families that are represented by members in the majority of the metazoans, providing a potential bridge to prevertebrate ancestral roles. This work investigates the structure and regulation of homologs of specific transcription factors known to regulate mammalian T and B cell development in a representative of the earliest diverging jawed vertebrates, the clearnose skate (Raja eglanteria). Skate orthologs of mammalian GATA-3, GATA-1, EBF-1, Pax-5, Pax-6, Runx2, and Runx3 have been characterized. GATA-3, Pax-5, Runx3, EBF-1, Spi-C, and most members of the Ikaros family are shown throughout ontogeny to be 1) coregulated with TCR or Ig expression, and 2) coexpressed with each other in combinations that for the most part correspond to known mouse T and B cell patterns, supporting conservation of function. These results indicate that multiple components of the gene regulatory networks that operate in mammalian T cell and B cell development were present in the common ancestor of the mammals and the cartilaginous fish. However, certain factors relevant to the B lineage differ in their tissue-specific expression patterns from their mouse counterparts, suggesting expanded or divergent B lineage characteristics or tissue specificity in these animals.
Collapse
Affiliation(s)
- Michele K Anderson
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
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
|
29
|
Carlsson R, Persson C, Leanderson T. SPI-C, a PU-box binding ETS protein expressed temporarily during B-cell development and in macrophages, contains an acidic transactivation domain located to the N-terminus. Mol Immunol 2003; 39:1035-43. [PMID: 12749910 DOI: 10.1016/s0161-5890(03)00032-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mice deficient for SPI-group ETS transcription factors PU.1 or SPI-B fail to generate lymphocytes or do not mount normal antibody mediated immune responses, respectively. PU.1 expression is restricted to B-, T-lymphocytes and macrophages, while SPI-B is expressed in B- and T-lymphocytes. SPI-C is an ETS transcription factor closely related to PU.1 and SPI-B, and expressed temporarily during B-cell development and in macrophages. By deletion and mutation analysis we show that the SPI-C protein has a transactivation domain located to the N-terminus, and that the transactivation activity is reduced to that of the DNA binding domain (DBD) alone when four aspartic acid residues are mutated to alanines. PU.1 and SPI-B regulate transcription from acidic domains located to the N-terminus and by recruiting the co-activator PIP to adjacent sites in a sequence specific manner. In contrast to PU.1 and PIP, SPI-C and PIP were unable to form a distinct ternary complex on the Ig lambda light chain lambda(2-4) enhancer element, suggesting that SPI-C could act both as a positive and negative transcriptional regulator during B-lymphocyte differentiation.
Collapse
Affiliation(s)
- Robert Carlsson
- Section for Immunology, CMB, Lund University, BMC I:13, 22184, Lund, Sweden.
| | | | | |
Collapse
|
30
|
Abstract
The Ets family of transcription factors characterized by an evolutionarily-conserved DNA-binding domain regulates expression of a variety of viral and cellular genes by binding to a purine-rich GGAA/T core sequence in cooperation with other transcriptional factors and co-factors. Most Ets family proteins are nuclear targets for activation of Ras-MAP kinase signaling pathway and some of them affect proliferation of cells by regulating the immediate early response genes and other growth-related genes. Some of them also regulate apoptosis-related genes. Several Ets family proteins are preferentially expressed in specific cell lineages and are involved in their development and differentiation by increasing the enhancer or promoter activities of the genes encoding growth factor receptors and integrin families specific for the cell lineages. Many Ets family proteins also modulate gene expression through protein-protein interactions with other cellular partners. Deregulated expression or formation of chimeric fusion proteins of Ets family due to proviral insertion or chromosome translocation is associated with leukemias and specific types of solid tumors. Several Ets family proteins also participate in malignancy of tumor cells including invasion and metastasis by activating the transcription of several protease genes and angiogenesis-related genes.
Collapse
Affiliation(s)
- Tsuneyuki Oikawa
- Department of Cell Genetics, Sasaki Institute, 2-2 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | | |
Collapse
|
31
|
Carlsson R, Hjalmarsson A, Liberg D, Persson C, Leanderson T. Genomic structure of mouse SPI-C and genomic structure and expression pattern of human SPI-C. Gene 2002; 299:271-8. [PMID: 12459275 DOI: 10.1016/s0378-1119(02)01078-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Erythroblast transformation-specific domain (ETS) transcription factors regulate some of the critical molecular mechanisms controlling the differentiation of multipotent haematopoietic progenitor cells into effector B-lymphocytes. The SPI-group ETS-protein transcription factors PU.1 and SPI-B play essential and, although coexpressed and binding to similar DNA sequences, unique roles in B-cell differentiation in mice. Mouse SPI-C is an SPI-group ETS protein expressed temporarily during B-cell development and in macrophages. Here we present the genomic organization of the mouse SPI-C gene, and show by rapid amplification of cDNA ends (5'-RACE) analysis that transcription of the mouse SPI-C mRNA starts at a single site producing a single processed transcript. We have also isolated a cDNA clone encoding the human SPI-C homologue, which displays 65% amino acid identity to the murine protein. In addition, we show that the genomic structure of the human and mouse genes are similar, containing a 5' non-coding exon followed by five coding exons. Human SPI-C mRNA is preferentially detected in foetal and adult spleen, lymph nodes and at lower levels in bone marrow and foetal liver. Finally a phylogenetic prediction analysis of SPI-group protein sequences suggest that the SPI-C proteins form a distinct subgroup, with human SPI-C being closest related to the mouse SPI-C protein.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Cloning, Molecular
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA-Binding Proteins/genetics
- Exons
- Gene Expression Profiling
- Genes/genetics
- HeLa Cells
- Humans
- Introns
- K562 Cells
- Mice
- Molecular Sequence Data
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- Transcription, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Robert Carlsson
- Section for Immunology, Department of Cell and Molecular Biology, BMC I:13, Lund University, 22184 Lund, Sweden.
| | | | | | | | | |
Collapse
|
32
|
Smith EMK, Gisler R, Sigvardsson M. Cloning and characterization of a promoter flanking the early B cell factor (EBF) gene indicates roles for E-proteins and autoregulation in the control of EBF expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:261-70. [PMID: 12077253 DOI: 10.4049/jimmunol.169.1.261] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The early B cell factor (EBF) is a transcription factor shown crucial for the development of B lymphocytes. The protein is expressed from the earliest stages of B cell development until the mature B cell stage, but the control elements responsible for the regulation of the gene are unknown. In this study, we report of the identification of a promoter region flanking the EBF gene. Several transcription start sites were identified by primer extension analysis in a region approximately 3.1 kb from the predicted ATG. Transient transfections revealed that this region was able to stimulate transcription of a reporter gene in B lymphoid and to a lesser extent, myeloid cells, but not in a pre-T cell line. The promoter was also able to functionally interact with E47, suggesting that the EBF gene may be a direct target for activation by E-proteins. In addition, functional binding of EBF to its own promoter was confirmed by EMSA and transfection assays indicating that the EBF protein may be involved in an autoregulatory loop. Finally, a tissue-restricted factor was able to bind an upstream regulatory region in B-lineage cells, further supporting the idea that the cloned promoter participates in the regulation of stage and lineage specific expression of the EBF gene.
Collapse
Affiliation(s)
- Emma M K Smith
- Laboratory of Cellular Differentiation, Department of Stem Cell Biology, Lund University, Lund, Sweden
| | | | | |
Collapse
|
33
|
Xuan Z, McCombie WR, Zhang MQ. GFScan: a gene family search tool at genomic DNA level. Genome Res 2002; 12:1142-9. [PMID: 12097353 PMCID: PMC186623 DOI: 10.1101/gr.220102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2001] [Accepted: 04/11/2002] [Indexed: 11/25/2022]
Abstract
We have developed GFScan(Gene Family Scan), a tool that identifies members of a gene family by searching genomic DNA sequences with genomic DNA motifs (or matrices) that are representative of the family. We have tested GFScan on four human gene families including the neurotransmitter-gated ion-channels (NGIC) family, the carbonic anhydrases (CA) family, the Dbl homology (DH) domain family, and the ETS-domain family. All known members of these families with motifs mapped to sequenced genomic DNA regions were found, whereas some novel genomic locations were also found to match the motifs, which may indicate new members in these families. Compared with other methods, GFScan recognized all true positives with much fewer false positives. We also showed that motifs constructed based on human genes could be used to search the mouse genome to identify orthologous family members in mouse. This program is available at http://www.cshl.org/mzhanglab/.
Collapse
Affiliation(s)
- Zhenyu Xuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | | | | |
Collapse
|
34
|
Gisler R, Sigvardsson M. The human V-preB promoter is a target for coordinated activation by early B cell factor and E47. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5130-8. [PMID: 11994467 DOI: 10.4049/jimmunol.168.10.5130] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of mature B lymphoid cells involves a highly orchestrated regulation of stage- and lineage-specific genes. In this study, we report an analysis of the human surrogate L chain VpreB promoter. The promoter has an overall homology of 56% to the mouse counterpart and displays a preB cell-restricted activity in transient transfections in cell lines. The promoter harbors three independent binding sites for early B cell factor (EBF) as defined by EMSA and supershift experiments. These sites were important for the full function of the promoter in a preB cell line, and chromatin immunoprecipitation experiments indicate that EBF interacts with the promoter in vivo. In addition to this, ectopic expression of EBF induces the activity of a reporter gene under control of the VpreB promoter in epithelioid HeLa cells, an effect augmented by coexpression of the basic-helix-loop helix transcription factor E47. The ability to interact directly with E47 was shared by the promoters controlling the human mb-1 and B29 genes. These data indicate that the human VpreB promoter is a direct target for activation by EBF and E47 and that functional collaboration between these proteins may be of great importance in human B cell development.
Collapse
Affiliation(s)
- Ramiro Gisler
- Laboratory for Cellular Differentiation, Department for Stem Cell Biology, BMC B12, 22184 Lund, Sweden.
| | | |
Collapse
|
35
|
Dahl R, Ramirez-Bergeron DL, Rao S, Simon M. Spi-B can functionally replace PU.1 in myeloid but not lymphoid development. EMBO J 2002; 21:2220-30. [PMID: 11980719 PMCID: PMC125373 DOI: 10.1093/emboj/21.9.2220] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2001] [Revised: 02/01/2002] [Accepted: 03/05/2002] [Indexed: 11/13/2022] Open
Abstract
Mature macrophages, neutrophils and lymphoid cells do not develop in PU.1(-/-) mice. In contrast, mice lacking the highly related protein Spi-B generate all hematopoietic lineages but display a B-cell receptor signaling defect. These distinct phenotypes could result from functional differences between PU.1 and Spi-B or their unique temporal and tissue-specific expression (PU.1: myeloid and B cells; Spi-B: B cells only). To address this question, we introduced the Spi-B cDNA into the murine PU.1 locus by homologous recombination. In the absence of PU.1, Spi-B rescued macrophage and granulocyte development when assayed by in vitro differentiation of embryonic stem cells. Adherent, CD11b(+)/F4/80(+) cells capable of phagocytosis were detected in PU.1(Spi-B/Spi-B) embryoid bodies, and myeloid colonies were present in hematopoietic progenitor assays. Despite its ability to rescue myeloid differentiation, Spi-B did not rescue lymphoid development in a RAG-2(-/-) complementation assay. These results demonstrate an important difference between PU.1 and Spi-B. Careful comparison of these Ets factors will delineate important functional domains of PU.1 involved in lymphocyte lineage commitment and/or maturation.
Collapse
Affiliation(s)
- Richard Dahl
- Abramson Family Cancer Research Institute and Howard Hughes Medical Institute, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104 and Department of Pathology, University of Chicago, Chicago, IL 60637, USA Corresponding author e-mail: R.Dahl and D.L.Ramirez-Bergeron contributed equally to this work
| | - Diana L. Ramirez-Bergeron
- Abramson Family Cancer Research Institute and Howard Hughes Medical Institute, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104 and Department of Pathology, University of Chicago, Chicago, IL 60637, USA Corresponding author e-mail: R.Dahl and D.L.Ramirez-Bergeron contributed equally to this work
| | - Sridhar Rao
- Abramson Family Cancer Research Institute and Howard Hughes Medical Institute, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104 and Department of Pathology, University of Chicago, Chicago, IL 60637, USA Corresponding author e-mail: R.Dahl and D.L.Ramirez-Bergeron contributed equally to this work
| | - M.Celeste Simon
- Abramson Family Cancer Research Institute and Howard Hughes Medical Institute, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104 and Department of Pathology, University of Chicago, Chicago, IL 60637, USA Corresponding author e-mail: R.Dahl and D.L.Ramirez-Bergeron contributed equally to this work
| |
Collapse
|
36
|
DeKoter RP, Lee HJ, Singh H. PU.1 regulates expression of the interleukin-7 receptor in lymphoid progenitors. Immunity 2002; 16:297-309. [PMID: 11869689 DOI: 10.1016/s1074-7613(02)00269-8] [Citation(s) in RCA: 230] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Development of the lymphoid system is dependent on the Ets family transcription factor PU.1. We demonstrate that PU.1(-/-) hematopoietic progenitors fail to express IL-7Ralpha transcripts. Promoter and chromatin crosslinking analyses suggest that PU.1 directly regulates transcription of the IL-7Ralpha gene. Retroviral transduction of IL-7Ralpha into PU.1(-/-) progenitors restores IL-7-dependent proliferation and induces, at low frequency, the generation of pro-B cells undergoing an apparently normal program of differentiation. Although the related factor Spi-B can substitute for PU.1 in early B cell development, it is not required. These results demonstrate that PU.1 functions to regulate early B cell development in part by controlling the expression of the IL-7Ralpha gene.
Collapse
Affiliation(s)
- Rodney P DeKoter
- Howard Hughes Medical Institute, Department of Molecular Genetics and Cell Biology, The University of Chicago, 5841 S. Maryland Ave., MC1028, Chicago, IL 60637, USA
| | | | | |
Collapse
|
37
|
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
|
38
|
Anderson KL, Nelson SL, Perkin HB, Smith KA, Klemsz MJ, Torbett BE. PU.1 is a lineage-specific regulator of tyrosine phosphatase CD45. J Biol Chem 2001; 276:7637-42. [PMID: 11114304 DOI: 10.1074/jbc.m009133200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The hematopoietic cell-specific ets family transcription factor PU.1 regulates many lymphoid and myeloid genes. We have determined that PU.1 is critical for lineage-specific expression of the tyrosine phosphatase CD45. CD45 is expressed exclusively in hematopoietic cells at all stages of development, except for mature red cells and platelets. Although CD45 is normally expressed in all leukocyte lineages, it is critically regulated by PU.1 only in myeloid cells. Whereas myeloid cells from PU.1 null mice failed to express CD45, lymphoid cells were CD45(+) by flow cytometry. Additionally, mRNA for CD45 was absent from PU.1-deficient myeloid cells. To understand the molecular basis for these observations, we characterized a transcriptional regulatory region of the murine CD45 gene containing exons 1a, 1b, and 2. Distinct transcriptional initiation sites for CD45 were demonstrated in T and B cells versus myeloid cells. A transcriptional initiation site in exon 1b (P1b) was principally utilized by myeloid cells. A PU.1 binding site was identified upstream of exon 1b by sequence analysis and DNA binding assays. Using this region of the CD45 locus we demonstrated that PU.1 directly transactivated reporter gene expression. Finally, retrovirus-mediated restoration of PU.1 expression to PU.1-deficient myeloid cells resulted in expression of cell surface CD45 and restored phosphatase activity, confirming the role of PU.1 in the positive regulation of this well known signaling molecule. We conclude that CD45 is regulated differentially in myeloid and lymphoid cells and that sequences critical to direct myeloid expression include a PU.1 binding site upstream of the P1b transcriptional initiation site.
Collapse
Affiliation(s)
- K L Anderson
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
39
|
Anderson MK, Sun X, Miracle AL, Litman GW, Rothenberg EV. Evolution of hematopoiesis: Three members of the PU.1 transcription factor family in a cartilaginous fish, Raja eglanteria. Proc Natl Acad Sci U S A 2001; 98:553-8. [PMID: 11149949 PMCID: PMC14625 DOI: 10.1073/pnas.98.2.553] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T lymphocytes and B lymphocytes are present in jawed vertebrates, including cartilaginous fishes, but not in jawless vertebrates or invertebrates. The origins of these lineages may be understood in terms of evolutionary changes in the structure and regulation of transcription factors that control lymphocyte development, such as PU.1. The identification and characterization of three members of the PU.1 family of transcription factors in a cartilaginous fish, Raja eglanteria, are described here. Two of these genes are orthologs of mammalian PU.1 and Spi-C, respectively, whereas the third gene, Spi-D, is a different family member. In addition, a PU.1-like gene has been identified in a jawless vertebrate, Petromyzon marinus (sea lamprey). Both DNA-binding and transactivation domains are highly conserved between mammalian and skate PU.1, in marked contrast to lamprey Spi, in which similarity is evident only in the DNA-binding domain. Phylogenetic analysis of sequence data suggests that the appearance of Spi-C may predate the divergence of the jawed and jawless vertebrates and that Spi-D arose before the divergence of the cartilaginous fish from the lineage leading to the mammals. The tissue-specific expression patterns of skate PU.1 and Spi-C suggest that these genes share regulatory as well as structural properties with their mammalian orthologs.
Collapse
Affiliation(s)
- M K Anderson
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | | | | | | |
Collapse
|
40
|
Evolution of hematopoiesis: Three members of the PU.1 transcription factor family in a cartilaginous fish, Raja eglanteria. Proc Natl Acad Sci U S A 2001. [PMID: 11149949 PMCID: PMC14625 DOI: 10.1073/pnas.021478998] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T lymphocytes and B lymphocytes are present in jawed vertebrates, including cartilaginous fishes, but not in jawless vertebrates or invertebrates. The origins of these lineages may be understood in terms of evolutionary changes in the structure and regulation of transcription factors that control lymphocyte development, such as PU.1. The identification and characterization of three members of the PU.1 family of transcription factors in a cartilaginous fish, Raja eglanteria, are described here. Two of these genes are orthologs of mammalian PU.1 and Spi-C, respectively, whereas the third gene, Spi-D, is a different family member. In addition, a PU.1-like gene has been identified in a jawless vertebrate, Petromyzon marinus (sea lamprey). Both DNA-binding and transactivation domains are highly conserved between mammalian and skate PU.1, in marked contrast to lamprey Spi, in which similarity is evident only in the DNA-binding domain. Phylogenetic analysis of sequence data suggests that the appearance of Spi-C may predate the divergence of the jawed and jawless vertebrates and that Spi-D arose before the divergence of the cartilaginous fish from the lineage leading to the mammals. The tissue-specific expression patterns of skate PU.1 and Spi-C suggest that these genes share regulatory as well as structural properties with their mammalian orthologs.
Collapse
|
41
|
Szymczyna BR, Arrowsmith CH. DNA binding specificity studies of four ETS proteins support an indirect read-out mechanism of protein-DNA recognition. J Biol Chem 2000; 275:28363-70. [PMID: 10867009 DOI: 10.1074/jbc.m004294200] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the ETS family of transcription factors are involved in several developmental and physiological processes, and, when overexpressed or misexpressed, can contribute to a variety of cancers. Each family member has a conserved DNA-binding domain that recognizes DNA sequences containing a G-G-A trinucleotide. Discrimination between potential ETS-binding sites appears to be governed by both the nucleotides flanking the G-G-A sequence and protein-protein interactions. We have used an adaptation of the "length-encoded multiplex" approach (Desjarlais, J. R., and Berg, J. M. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 11099-11103) to define DNA binding specificities for four ETS proteins: Fli-1, SAP-1, PU.1, and TEL. Our results support a model in which cooperative effects among neighboring bases flanking the central G-G-A site contribute to the formation of stable ETS/DNA complexes. These results are consistent with a mechanism for specific DNA binding that is partially governed by an indirect read-out of the DNA sequence, in which a sequence-specific DNA conformation is sensed or induced.
Collapse
Affiliation(s)
- B R Szymczyna
- Ontario Cancer Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | | |
Collapse
|
42
|
Shintani S, Terzic J, Sato A, Saraga-Babic M, O'hUigin C, Tichy H, Klein J. Do lampreys have lymphocytes? The Spi evidence. Proc Natl Acad Sci U S A 2000; 97:7417-22. [PMID: 10840049 PMCID: PMC16560 DOI: 10.1073/pnas.110505597] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It is generally accepted that living jawless vertebrates (lampreys and hagfishes) lack the capability of mounting an adaptive immune response. At the same time, however, there are reports describing histological evidence for the presence in agnathan tissues of lymphocytes, the key players in adaptive immunity. The question therefore arises whether the cells identified morphologically as lymphocytes are true lymphocytes in terms of their genetic developmental program. In this study, evidence is provided that the lampreys express a member of the purine box 1 (PU.1)/spleen focus-forming virus integration B (Spi-B) gene family known to be critically and specifically involved in the differentiation of lymphocytes in jawed vertebrates. The lamprey gene is expressed in the lymphocyte-like cells of the digestive tract and inexplicably also in the ovary.
Collapse
Affiliation(s)
- S Shintani
- Max-Planck-Institut für Biologie, Abteilung Immungenetik, Correnstrasse 42, D-7076 Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Ray-Gallet D, Moreau-Gachelin F. Phosphorylation of the Spi-B transcription factor reduces its intrinsic stability. FEBS Lett 1999; 464:164-8. [PMID: 10618498 DOI: 10.1016/s0014-5793(99)01687-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Spi-B transcription factor is an Ets protein expressed in B lymphoid cells and closely related to the Spi-1/PU.1 oncoprotein. By mutational analysis, we showed that Spi-B is phosphorylated by casein kinase II in vitro on four serine residues. Mutation of these four serines to alanines prevented the phosphorylation of Spi-B in vivo, increased the ability of Spi-B to transactivate expression of a reporter gene and led to a decrease of Spi-B stability. We propose that the phosphorylation of Spi-B may participate in the modulation of Spi-B functional activity by controlling its intracellular protein level.
Collapse
Affiliation(s)
- D Ray-Gallet
- INSERM, Unité 248, Institut Curie, 26 rue díUlm, 75248, Paris, France
| | | |
Collapse
|
44
|
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
|