101
|
The transcription factor Gfi1 regulates G-CSF signaling and neutrophil development through the Ras activator RasGRP1. Blood 2010; 115:3970-9. [PMID: 20203268 DOI: 10.1182/blood-2009-10-246967] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The transcription factor growth factor independence 1 (Gfi1) and the growth factor granulocyte colony-stimulating factor (G-CSF) are individually essential for neutrophil differentiation from myeloid progenitors. Here, we provide evidence that the functions of Gfi1 and G-CSF are linked in the regulation of granulopoiesis. We report that Gfi1 promotes the expression of Ras guanine nucleotide releasing protein 1 (RasGRP1), an exchange factor that activates Ras, and that RasGRP1 is required for G-CSF signaling through the Ras/mitogen-activated protein/extracellular signal-regulated kinase (MEK/Erk) pathway. Gfi1-null mice have reduced levels of RasGRP1 mRNA and protein in thymus, spleen, and bone marrow, and Gfi1 transduction in myeloid cells promotes RasGRP1 expression. When stimulated with G-CSF, Gfi1-null myeloid cells are selectively defective at activating Erk1/2, but not signal transducer and activator of transcription 1 (STAT1) or STAT3, and fail to differentiate into neutrophils. Expression of RasGRP1 in Gfi1-deficient cells rescues Erk1/2 activation by G-CSF and allows neutrophil maturation by G-CSF. These results uncover a previously unknown function of Gfi1 as a regulator of RasGRP1 and link Gfi1 transcriptional control to G-CSF signaling and regulation of granulopoiesis.
Collapse
|
102
|
Barjaktarevic I, Maletkovic-Barjaktarevic J, Kamani NR, Vukmanovic S. Altered functional balance of Gfi-1 and Gfi-1b as an alternative cause of reticular dysgenesis? Med Hypotheses 2010; 74:445-8. [DOI: 10.1016/j.mehy.2009.09.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 09/27/2009] [Indexed: 12/20/2022]
|
103
|
Laurent B, Randrianarison-Huetz V, Kadri Z, Roméo PH, Porteu F, Duménil D. Gfi-1B promoter remains associated with active chromatin marks throughout erythroid differentiation of human primary progenitor cells. Stem Cells 2009; 27:2153-62. [PMID: 19522008 PMCID: PMC2962905 DOI: 10.1002/stem.151] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Growth Factor Independent-1B (Gfi-1B) is a transcriptional repressor that plays critical roles in the control of erythropoiesis and megakaryopoiesis. Gfi-1B expression was described to be repressed by an autoregulatory feedback control loop. Here, we show that Gfi-1 transcription is positively regulated early after induction of erythroid differentiation and remains highly active to late erythroblasts. Using chromatin immunoprecipitation assays in CD34+ cells from human cord blood, we found that Gfi-1 and GATA-2 in immature progenitors and then Gfi-1B and GATA-1 in erythroblasts are bound to the Gfi-1B promoter as well as to the promoter of c-myc, a known Gfi-1B target gene. Surprisingly, this Gfi-1/GATA-2–Gfi-1B/GATA-1 switch observed at erythroblast stages is associated to an increase in the Gfi-1B transcription whereas it triggers repression of c-myc transcription. Accordingly, analysis of chromatin modification patterns shows that HDAC, CoREST, and LSD1 are recruited to the c-myc promoter leading to appearance of repressive chromatin marks. In contrast, the Gfi-1B promoter remains associated with a transcriptionally active chromatin configuration as highlighted by an increase in histone H3 acetylation and concomitant release of the LSD1 and CoREST corepressors. The repressive function of Gfi-1B therefore depends on the nature of the proteins recruited to the target gene promoters and on chromatin modifications. We conclude that Gfi-1B behaves as a lineage-affiliated gene with an open chromatin configuration in multipotent progenitors and sustained activation as cells progress throughout erythroid differentiation.
Collapse
Affiliation(s)
- Benoît Laurent
- Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (UMR 8104), Paris, France
| | | | | | | | | | | |
Collapse
|
104
|
High-mobility group protein HMGB2 regulates human erythroid differentiation through trans-activation of GFI1B transcription. Blood 2009; 115:687-95. [PMID: 19965638 DOI: 10.1182/blood-2009-06-230094] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gfi-1B is a transcriptional repressor that is crucial for erythroid differentiation: inactivation of the GFI1B gene in mice leads to embryonic death due to failure to produce differentiated red cells. Accordingly, GFI1B expression is tightly regulated during erythropoiesis, but the mechanisms involved in such regulation remain partially understood. We here identify HMGB2, a high-mobility group HMG protein, as a key regulator of GFI1B transcription. HMGB2 binds to the GFI1B promoter in vivo and up-regulates its trans-activation most likely by enhancing the binding of Oct-1 and, to a lesser extent, of GATA-1 and NF-Y to the GFI1B promoter. HMGB2 expression increases during erythroid differentiation concomitantly to the increase of GfI1B transcription. Importantly, knockdown of HMGB2 in immature hematopoietic progenitor cells leads to decreased Gfi-1B expression and impairs their erythroid differentiation. We propose that HMGB2 potentiates GATA-1-dependent transcription of GFI1B by Oct-1 and thereby controls erythroid differentiation.
Collapse
|
105
|
Anguita E, Villegas A, Iborra F, Hernández A. GFI1B controls its own expression binding to multiple sites. Haematologica 2009; 95:36-46. [PMID: 19773260 DOI: 10.3324/haematol.2009.012351] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Transcription factors play essential roles in both normal and malignant hematopoiesis. This is the case for the growth factor independent 1b (GFI1B) transcription factor, which is required for erythroid and megakaryocytic differentiation and over-expressed in leukemic patients and cell lines. DESIGN AND METHODS To investigate GFI1B regulation, we searched for multispecies conserved non-coding elements between GFI1B and neighboring genes. We used a formaldehyde-assisted isolation of regulatory elements (FAIRE) assay and DNase1 hypersensitivity to assess the chromatin conformation of these sites. Next, we analyzed transcription factor binding and histone modifications at the GFI1B locus including the conserved non-coding elements by a chromatin immunoprecipitation assay. Finally, we studied the interaction of the GFI1B promoter and the conserved non-coding elements with the chromatin conformation capture technique and used immunofluorescence to evaluate GFI1B levels in individual cells. RESULTS We localized several conserved non-coding elements containing multiple erythroid specific transcription factor binding sites at the GFI1B locus. In GFI1B-expressing cells a subset of these conserved non-coding elements and the promoter adopt a close spatial conformation, localize with open chromatin sites, harbor chromatin modifications associated with gene activation and bind multiple transcription factors and co-repressors. Conclusions Our findings indicate that GFI1B regulatory elements behave as activators and repressors. Different protein levels within a cell population suggest that cells must activate and repress GFI1B continuously to control its final level. These data are consistent with a model of GFI1B regulation in which GFI1B binds to its own promoter and to the conserved non-coding elements as its levels rise. This would attract repressor complexes that progressively down-regulate the gene. GFI1B expression would decrease until a stage at which the activating complexes predominate and expression increases.
Collapse
Affiliation(s)
- Eduardo Anguita
- Hematology Department, Hospital Clinico San Carlos, 28040 Madrid, Spain.
| | | | | | | |
Collapse
|
106
|
Ichiyama K, Hashimoto M, Sekiya T, Nakagawa R, Wakabayashi Y, Sugiyama Y, Komai K, Saba I, Moroy T, Yoshimura A. Gfi1 negatively regulates Th17 differentiation by inhibiting ROR t activity. Int Immunol 2009; 21:881-9. [DOI: 10.1093/intimm/dxp054] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
107
|
Vassen L, Khandanpour C, Ebeling P, van der Reijden BA, Jansen JH, Mahlmann S, Dührsen U, Möröy T. Growth factor independent 1b (Gfi1b) and a new splice variant of Gfi1b are highly expressed in patients with acute and chronic leukemia. Int J Hematol 2009; 89:422-430. [PMID: 19360458 DOI: 10.1007/s12185-009-0286-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 01/23/2009] [Accepted: 02/23/2009] [Indexed: 10/20/2022]
Abstract
Gfi1b is a transcriptional repressor that is essential for erythroid cells and megakaryocytes, but is also expressed in hematopoietic stem cells and early myeloid progenitors. The chromosomal localization of the Gfi1b gene at 9q34 and its functional homology with the proto-oncogene Gfi1 were suggestive for a role of Gfi1b in malignant transformation and myeloid leukemia. We show here that the expression of Gfi1b is strongly elevated in CML and AML patients compared to normal healthy controls and that imatinib, a drug widely used to treat CML, further enhances Gfi1b expression in patients even after remission. Our data suggest that Gfi1b may be an important factor to establish or maintain myeloid leukemia and myeloproliferative diseases and that, high expression levels of Gfi1b might be associated with the emergence of Philadelphia chromosome negative myeloid malignancies after imatinib withdrawal or after the development of imatinib resistance.
Collapse
Affiliation(s)
- Lothar Vassen
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Virchowstrasse 173, 45122, Essen, Germany.,Institut de recherches cliniques de Montreal, IRCM, 110 Avenue des Pins West, Montreal, QC, H2W 1R7, Canada
| | - Cyrus Khandanpour
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Virchowstrasse 173, 45122, Essen, Germany.,Institut de recherches cliniques de Montreal, IRCM, 110 Avenue des Pins West, Montreal, QC, H2W 1R7, Canada
| | - Peter Ebeling
- Department for Internal Medicine, Center for Cancer Research, Universitätsklinikum Essen, 45122, Essen, Germany
| | - Bert A van der Reijden
- Central Hematology Laboratory, Radboud University Nijmegen Medical Centre for Molecular Life Sciences (NCMLS), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Joop H Jansen
- Central Hematology Laboratory, Radboud University Nijmegen Medical Centre for Molecular Life Sciences (NCMLS), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Stefan Mahlmann
- Department of Internal Medicine and Hematology, Universitätsklinikum Essen, 45122, Essen, Germany
| | - Ulrich Dührsen
- Department of Internal Medicine and Hematology, Universitätsklinikum Essen, 45122, Essen, Germany
| | - Tarik Möröy
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Virchowstrasse 173, 45122, Essen, Germany. .,Institut de recherches cliniques de Montreal, IRCM, 110 Avenue des Pins West, Montreal, QC, H2W 1R7, Canada. .,Departement de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada.
| |
Collapse
|
108
|
Abstract
In patients with severe congenital neutropenia (SCN) and mice with growth factor independent-1 (Gfi1) loss of function, arrested myeloid progenitors accumulate, whereas terminal granulopoiesis is blocked. One might assume that Gfi-null progenitors accumulate because they lack the ability to differentiate. Instead, our data indicate that Gfi1 loss of function deregulates 2 separable transcriptional programs, one of which controls the accumulation and lineage specification of myeloid progenitors, but not terminal granulopoiesis. We demonstrate that Gfi1 directly represses HoxA9, Pbx1, and Meis1 during normal myelopoiesis. Gfi1-/- progenitors exhibit elevated levels of HoxA9, Pbx1 and Meis1, exaggerated HoxA9-Pbx1-Meis1 activity, and progenitor transformation in collaboration with oncogenic K-Ras. Limiting HoxA9 alleles corrects, in a dose-dependent manner, in vivo and in vitro phenotypes observed with loss of Gfi1 in myeloid progenitor cells but did not rescue Gfi1-/- blocked granulopoiesis. Thus, Gfi1 integrates 2 events during normal myeloid differentiation; the suppression of a HoxA9-Pbx1-Meis1 progenitor program and the induction of a granulopoietic transcription program.
Collapse
|
109
|
Loss of the transcriptional repressor PAG-3/Gfi-1 results in enhanced neurosecretion that is dependent on the dense-core vesicle membrane protein IDA-1/IA-2. PLoS Genet 2009; 5:e1000447. [PMID: 19343207 PMCID: PMC2657203 DOI: 10.1371/journal.pgen.1000447] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 03/06/2009] [Indexed: 01/09/2023] Open
Abstract
It is generally accepted that neuroendocrine cells regulate dense core vesicle (DCV) biogenesis and cargo packaging in response to secretory demands, although the molecular mechanisms of this process are poorly understood. One factor that has previously been implicated in DCV regulation is IA-2, a catalytically inactive protein phosphatase present in DCV membranes. Our ability to directly visualize a functional, GFP-tagged version of an IA-2 homolog in live Caenorhabditis elegans animals has allowed us to capitalize on the genetics of the system to screen for mutations that disrupt DCV regulation. We found that loss of activity in the transcription factor PAG-3/Gfi-1, which functions as a repressor in many systems, results in a dramatic up-regulation of IDA-1/IA-2 and other DCV proteins. The up-regulation of DCV components was accompanied by an increase in presynaptic DCV numbers and resulted in phenotypes consistent with increased neuroendocrine secretion. Double mutant combinations revealed that these PAG-3 mutant phenotypes were dependent on wild type IDA-1 function. Our results support a model in which IDA-1/IA-2 is a critical element in DCV regulation and reveal a novel genetic link to PAG-3-mediated transcriptional regulation. To our knowledge, this is the first mutation identified that results in increased neurosecretion, a phenotype that has clinical implications for DCV-mediated secretory disorders. Within secretory cells, hormones are packaged into vesicles (called DCVs) that are released upon stimulation. The number of DCVs is regulated to meet the secretory demands of the cell by a mechanism that is poorly understood, although a protein in the membrane of DCVs, called IA-2, is thought to play a role. A genetic screen in the nematode C. elegans is used, here, to find mutations that mis-regulate the corresponding worm protein called IDA-1. Capitalizing on the simple neuroanatomy of the nematode and its transparency, we visualize IDA-1 protein levels directly in the animal using a fluorescent tag. We find that mutations in the transcription factor PAG-3/Gfi-1 result in elevated levels of IDA-1 protein, increased numbers of presynaptic DCVs, and behaviors consistent with increased neurosecretion. Our results demonstrate that IDA-1/IA-2 protein levels correlate with the biogenesis, utilization, or stability of DCVs. We propose that PAG-3 normally down regulates the production of IDA-1, thus serving as part of the mechanism underlying DCV regulation. This is the first reported mutation that increases DCV numbers and secretion, offering insight into DCV homeostasis and a potential therapeutic target for diseases that would benefit from a boost in neuroendocrine secretion.
Collapse
|
110
|
Zhu J, Davidson TS, Wei G, Jankovic D, Cui K, Schones DE, Guo L, Zhao K, Shevach EM, Paul WE. Down-regulation of Gfi-1 expression by TGF-beta is important for differentiation of Th17 and CD103+ inducible regulatory T cells. ACTA ACUST UNITED AC 2009; 206:329-41. [PMID: 19188499 PMCID: PMC2646571 DOI: 10.1084/jem.20081666] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Growth factor independent 1 (Gfi-1), a transcriptional repressor, is transiently induced during T cell activation. Interleukin (IL) 4 further induces Gfi-1, resulting in optimal Th2 cell expansion. We report a second important function of Gfi-1 in CD4 T cells: prevention of alternative differentiation by Th2 cells, and inhibition of differentiation of naive CD4 T cells to either Th17 or inducible regulatory T (iTreg) cells. In Gfi1−/− Th2 cells, the Rorc, Il23r, and Cd103 loci showed histone 3 lysine 4 trimethylation modifications that were lacking in wild-type Th2 cells, implying that Gfi-1 is critical for epigenetic regulation of Th17 and iTreg cell–related genes in Th2 cells. Enforced Gfi-1 expression inhibited IL-17 production and iTreg cell differentiation. Furthermore, a key inducer of both Th17 and iTreg cell differentiation, transforming growth factor β, repressed Gfi-1 expression, implying a reciprocal negative regulation of CD4 T cell fate determination. Chromatin immunoprecipitation showed direct binding of the Gfi-1–lysine-specific demethylase 1 repressive complex to the intergenic region of Il17a/Il17f loci and to intron 1 of Cd103. T cell–specific Gfi1 conditional knockout mice displayed a striking delay in the onset of experimental allergic encephalitis correlated with a dramatic increase of Foxp3+CD103+ CD4 T cells. Thus, Gfi-1 plays a critical role both in enhancing Th2 cell expansion and in repressing induction of Th17 and CD103+ iTreg cells.
Collapse
Affiliation(s)
- Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Igwe E, Kosan C, Khandanpour C, Sharif-Askari E, Brüne B, Möröy T. The zinc finger protein Gfi1 is implicated in the regulation of IgG2b production and the expression of Igamma2b germline transcripts. Eur J Immunol 2009; 38:3004-14. [PMID: 18991277 DOI: 10.1002/eji.200838251] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gfi1 is a zinc finger transcription factor that is undetectable in B lymphocytes but its expression rises rapidly upon antigenic stimulation or treatment with lipopolysaccharide (LPS). Here we show that Gfi1(-/-) mice have higher serum levels of gamma isotype immunoglobulin than WT animals. When challenged with antigen, Gfi1(-/-) mice react with accelerated formation of PNA+/CD19+ germinal center B cells and an increased production of antigen-specific IgG2a and IgG2b. Moreover, Gfi1(-/-) B cells secrete more IgG2a and IgG2b than WT cells and produce higher levels of Igamma2b sterile germline transcripts when cultured with LPS. While the proliferative response to stimulation with anti-IgM antibodies and plasma cell differentiation was normal in Gfi1(-/-) B cells, we found that mRNA and protein levels of TGFbeta1 were significantly increased in the absence of Gfi1. TGFbeta1 has been shown to be essential for the regulation of IgG subclass production and was previously found to selectively stimulate IgG2b secretion. Our findings reveal a new function of Gfi1 in the control of IgG isotype production.
Collapse
Affiliation(s)
- Emeka Igwe
- Institut für Zellbiologie Tumorforschung, IFZ, Universitätsklinikum Essen, Essen, Germany.
| | | | | | | | | | | |
Collapse
|
112
|
Li W, Ma N, Ong LL, Kaminski A, Skrabal C, Ugurlucan M, Lorenz P, Gatzen HH, Lützow K, Lendlein A, Pützer BM, Li RK, Steinhoff G. Enhanced thoracic gene delivery by magnetic nanobead-mediated vector. J Gene Med 2008; 10:897-909. [PMID: 18481827 DOI: 10.1002/jgm.1208] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Systemic gene delivery is limited by the adverse hydrodynamic conditions on the collection of gene carrier particles to the specific area. In the present study, a magnetic field was employed to guide magnetic nanobead (MNB)/polymer/DNA complexes after systemic administration to the left side of the mouse thorax in order to induce localized gene expression. METHODS Nonviral polymer (poly ethyleneimine, PEI) vector-gene complexes were conjugated to MNBs with the Sulfo-NHS-LC-Biotin linker. In vitro transfection efficacy of MNB/PEI/DNA was compared with PEI/DNA in three different cell lines as well as primary endothelial cells under magnetic field stimulation. In vivo, MNB/PEI/DNA complexes were injected into the tail vein of mice and an epicardial magnet was employed to attract the circulating MNB/PEI/DNA complexes. RESULTS Endocytotic uptake of MNB/PEI/DNA complexes and intracellular gene release with nuclear translocation were observed in vitro, whereas the residues of MNB/PEI complexes were localized at the perinuclear region. Compared with PEI/DNA complexes alone, MNB/PEI/DNA complexes had a 36- to 85-fold higher transfection efficiency under the magnetic field. In vivo, the epicardial magnet effectively attracted MNB/PEI/DNA complexes in the left side of the thorax, resulting in strong reporter and therapeutic gene expression in the left lung and the heart. Gene expression in the heart was mainly within the endothelium. CONCLUSIONS MNB-mediated gene delivery could comprise a promising method for gene delivery to the lung and the heart.
Collapse
Affiliation(s)
- Wenzhong Li
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Montoya-Durango DE, Velu CS, Kazanjian A, Rojas MEB, Jay CM, Longmore GD, Grimes HL. Ajuba functions as a histone deacetylase-dependent co-repressor for autoregulation of the growth factor-independent-1 transcription factor. J Biol Chem 2008; 283:32056-65. [PMID: 18805794 DOI: 10.1074/jbc.m802320200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth factor independent-1 (Gfi1) is a zinc finger protein with a SNAG-transcriptional repressor domain. Ajuba is a LIM domain protein that shuttles between the cytoplasm and the nucleus. Ajuba functions as a co-repressor for synthetic Gfi1 SNAG-repressor domain-containing constructs, but a role for Ajuba co-repression of the cognate DNA bound Gfi1 protein has not been defined. Co-immunoprecipitation of synthetic and endogenous proteins and co-elution with gel filtration suggest that an endogenous Ajuba.Gfi1.HDAC multiprotein complex is possible. Active histone deacetylase activity co-immunoprecipitates with Ajuba or Gfi1, and both proteins depend upon histone deacetylases for full transcriptional repression activity. Ajuba LIM domains directly bind to Gfi1, but the association is not SNAG domain-dependent. ChIP analysis and reciprocal knockdown experiments suggest that Ajuba selectively functions as a co-repressor for Gfi1 autoregulation. The data suggest that Ajuba is utilized as a corepressor selectively on Gfi1 target genes.
Collapse
Affiliation(s)
- Diego E Montoya-Durango
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
114
|
Li-Kroeger D, Witt LM, Grimes HL, Cook TA, Gebelein B. Hox and senseless antagonism functions as a molecular switch to regulate EGF secretion in the Drosophila PNS. Dev Cell 2008; 15:298-308. [PMID: 18694568 DOI: 10.1016/j.devcel.2008.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 05/08/2008] [Accepted: 06/04/2008] [Indexed: 01/27/2023]
Abstract
Hox factors are key regulators of distinct cells, tissues, and organs along the body plan. However, little is known about how Hox factors regulate cell-specific gene expression to pattern diverse tissues. Here, we show an unexpected Hox transcriptional mechanism: the permissive regulation of EGF secretion, and thereby cell specification, by antagonizing the Senseless transcription factor in the peripheral nervous system. rhomboid expression in a subset of sensory cells stimulates EGF secretion to induce hepatocyte-like cell development. We identified a rhomboid enhancer that is active in these cells and show that an abdominal Hox complex directly competes with Senseless for enhancer binding, with the transcriptional outcome dependent upon their relative binding activities. Thus, Hox-Senseless antagonism forms a molecular switch that integrates neural and anterior-posterior positional information. As the vertebrate senseless homolog is essential for neural development as well as hematopoiesis, we propose Hox-Senseless antagonism will broadly control cell fate decisions.
Collapse
Affiliation(s)
- David Li-Kroeger
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
115
|
Shinnakasu R, Yamashita M, Kuwahara M, Hosokawa H, Hasegawa A, Motohashi S, Nakayama T. Gfi1-mediated stabilization of GATA3 protein is required for Th2 cell differentiation. J Biol Chem 2008; 283:28216-25. [PMID: 18701459 DOI: 10.1074/jbc.m804174200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The differentiation of naive CD4 T cells into Th2 cells requires the T cell receptor-mediated activation of the ERK MAPK cascade. Little is known, however, in regard to how the ERK MAPK cascade regulates Th2 cell differentiation. We herein identified Gfi1 (growth factor independent-1) as a downstream target of the ERK MAPK cascade for Th2 cell differentiation. In the absence of Gfi1, interleukin-5 production and the change of histone modification at the interleukin-5 gene locus were severely impaired. Furthermore, the interferon gamma gene showed a striking activation in the Gfi1(-/-) Th2 cells. An enhanced ubiquitin/proteasome-dependent degradation of GATA3 protein was observed in Gfi1(-/-) Th2 cells, and the overexpression of GATA3 eliminated the defect of Th2 cell function in Gfi1-deficient Th2 cells. These data suggest that the T cell receptor-mediated induction of Gfi1 controls Th2 cell differentiation through the regulation of GATA3 protein stability.
Collapse
Affiliation(s)
- Ryo Shinnakasu
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
116
|
Gfi1.1 regulates hematopoietic lineage differentiation during zebrafish embryogenesis. Cell Res 2008; 18:677-85. [PMID: 18504458 DOI: 10.1038/cr.2008.60] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Growth factor independence 1 (GFI1) is important for maturation of mammalian lymphocytes and neutrophils and maintenance of adult hematopoietic stem cells (HSCs). The role of GFI1 in embryonic hematopoiesis is less well characterized. Through an enhancer trap screen and bioinformatics analysis, we identified a zebrafish homolog of Gfi1 (named gfi1.1) and analyzed its function during embryonic development. Expression of both an endogenous gfi1.1 gene and a GFP reporter gene inserted near its genomic locus was detected in hematopoietic cells of zebrafish embryos. Morpholino (MO) knockdown of gfi1.1 reduced expression of scl, lmo2, c-myb, mpo, rag1, gata1 and hemoglobin alpha embryonic-1 (hbae1), as well as the total amount of embryonic hemoglobin, but increased expression of pu.1 and l-plastin. Under the same conditions, MO injection did not affect the markers involved in vascular and pronephric development. Conversely, overexpression of gfi1.1 via mRNA injection enhanced expression of gata1 but inhibited expression of pu.1. These findings suggest that Gfi1.1 plays a critical role in regulating the balance of embryonic erythroid and myeloid lineage determination, and is also required for the differentiation of lymphocytes and granulocytes during zebrafish embryogenesis.
Collapse
|
117
|
Prox1 interacts with Atoh1 and Gfi1, and regulates cellular differentiation in the inner ear sensory epithelia. Dev Biol 2008; 322:33-45. [PMID: 18652815 DOI: 10.1016/j.ydbio.2008.07.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Revised: 06/28/2008] [Accepted: 07/01/2008] [Indexed: 01/14/2023]
Abstract
Inner ear hair cells and supporting cells arise from common precursors and, in mammals, do not show phenotypic conversion. Here, we studied the role of the homeodomain transcription factor Prox1 in the inner ear sensory epithelia. Adenoviral-mediated Prox1 transduction into hair cells in explant cultures led to strong repression of Atoh1 and Gfi1, two transcription factors critical for hair cell differentiation and survival. Luciferase assays showed that Prox1 can repress transcriptional activity of Gfi1 independently of Atoh1. Prox1 transduction into cochlear outer hair cells resulted in degeneration of these cells, consistent with the known phenotype of Gfi1-deficient mice. These results together with the widespread expression of endogenous Prox1 within the population of inner ear supporting cells point to the role for Prox1 in antagonizing the hair cell phenotype in these non-sensory cells. Further, in vivo analyses of hair cells from Gfi1-deficient mice suggest that the cyclin-dependent kinase inhibitor p57(Kip2) mediates the differentiation- and survival-promoting functions of Gfi1. These data reveal novel gene interactions and show that these interactions regulate cellular differentiation within the inner ear sensory epithelia. The data point to the tight regulation of phenotypic characteristics of hair cells and supporting cells.
Collapse
|
118
|
Stern JL, Cao JZ, Xu J, Mocarski ES, Slobedman B. Repression of human cytomegalovirus major immediate early gene expression by the cellular transcription factor CCAAT displacement protein. Virology 2008; 378:214-25. [PMID: 18614194 DOI: 10.1016/j.virol.2008.05.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 05/13/2008] [Accepted: 05/22/2008] [Indexed: 01/19/2023]
Abstract
Initiation of human cytomegalovirus (HCMV) productive infection is dependent on the major immediate early (MIE) genes ie1 and ie2. Several putative binding sites for CCAAT displacement protein (CDP or CUX1) were identified within the MIE promoter/regulatory region. Binding assays demonstrated binding of CUX1 to MIE-region oligonucleotides containing the CUX1 core binding sequence ATCGAT and mutagenesis of this sequence abrogated CUX1 binding. Furthermore, CUX1 repressed expression of a luciferase reporter construct controlled by the MIE promoter, and mutation of CUX1 binding sites within the promoter diminished this repressive function of CUX1. In the context of virus infection of HEK293 cells transfected with the CUX1 expression vector, CUX1 showed evidence of association with the HCMV MIE regulatory region and inhibited the capacity of the virus to express ie1 and ie2 transcripts, suggesting that this cellular factor regulates MIE gene expression following virus entry. These data identify a role for CUX1 in repressing HCMV gene expression essential for initiation of the replicative cycle.
Collapse
Affiliation(s)
- J Lewis Stern
- Centre for Virus Research, Westmead Millennium Institute, PO Box 412, Westmead, New South Wales 2145, Australia
| | | | | | | | | |
Collapse
|
119
|
Chandele A, Joshi NS, Zhu J, Paul WE, Leonard WJ, Kaech SM. Formation of IL-7Ralphahigh and IL-7Ralphalow CD8 T cells during infection is regulated by the opposing functions of GABPalpha and Gfi-1. THE JOURNAL OF IMMUNOLOGY 2008; 180:5309-19. [PMID: 18390712 DOI: 10.4049/jimmunol.180.8.5309] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IL-7 is essential for the survival of naive and memory T cells, and IL-7 receptor alpha-chain (IL-7Ralpha) expression is dynamically regulated in activated CD8 T cells during acute viral and bacterial infections. Most virus-specific CD8 T cells become IL-7Ralpha(low) and are relatively short-lived, but some escape IL-7Ralpha repression (referred to as IL-7Ralpha(high) memory precursor effector cells) and preferentially enter the memory CD8 T cell pool. How antiviral effector CD8 T cells regulate IL-7Ralpha expression in an "on and off" fashion remains to be characterized. During lymphocytic choriomeningitis virus infection, we found that opposing actions of the transcription factors GABPalpha (GA binding protein alpha) and Gfi-1 (growth factor independence 1) control IL-7Ralpha expression in effector CD8 T cells. Specifically, GABPalpha was required for IL-7Ralpha expression in memory precursor effector cells, and this correlated with hyperacetylation of the Il7ra promoter. In contrast, Gfi-1 was required for stable IL-7Ralpha repression in effector CD8 T cells and acted by antagonizing GABPalpha binding and recruiting histone deacetylase 1, which deacetylated the Il7ra promoter. Thus, Il7ra promoter acetylation and activity was dependent on the reciprocal binding of GABPalpha and Gfi-1, and these data provide a biochemical mechanism for the generation of stable IL-7Ralpha(high) and IL-7Ralpha(low) states in virus-specific effector CD8 T cells.
Collapse
Affiliation(s)
- Anmol Chandele
- Department of Immunobiology, Yale Medical School, New Haven, CT 06511, USA
| | | | | | | | | | | |
Collapse
|
120
|
Fink MP. Bench-to-bedside review: High-mobility group box 1 and critical illness. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 11:229. [PMID: 17903310 PMCID: PMC2556731 DOI: 10.1186/cc6088] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
High-mobility group box 1 (HMGB1) is a DNA-binding protein that also exhibits proinflammatory cytokine-like activity. HMGB1 is passively released by necrotic cells and also is actively secreted by immunostimulated macrophages, dendritic cells, and enterocytes. Although circulating HMGB1 levels are increased relative to healthy controls in patients with infections and severe sepsis, plasma or serum HMGB1 concentrations do not discriminate reliably between infected and uninfected critically ill patients. Nevertheless, administration of drugs that block HMGB1 secretion or of anti-HMGB1 neutralizing antibodies has been shown to ameliorate organ dysfunction and/or improve survival in numerous animal models of critical illness. Because HMGB1 tends to be released relatively late in the inflammatory response (at least in animal models of endotoxemia or sepsis), this protein is an attractive target for the development of new therapeutic agents for the treatment of patients with various forms of critical illness.
Collapse
Affiliation(s)
- Mitchell P Fink
- Department of Critical Care Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
121
|
Sinclair J. Human cytomegalovirus: Latency and reactivation in the myeloid lineage. J Clin Virol 2008; 41:180-5. [PMID: 18164651 DOI: 10.1016/j.jcv.2007.11.014] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 11/07/2007] [Accepted: 11/09/2007] [Indexed: 02/08/2023]
Abstract
Human cytomegalovirus (HCMV) persists as a sub-clinical, lifelong infection in the human host which is maintained at least in part by its carriage in the absence of detectable infectious virus: a hallmark of latent infection. In contrast, reactivation from latency in immuno-compromised individuals can result in serious disease. Understanding virus latency and reactivation, therefore, is essential for a full understanding of the biology and pathogenesis of this persistent human herpesvirus. However, the precise cellular sites in which HCMV is carried and the mechanisms regulating its latency and reactivation, during natural infection, remain poorly understood. Recent work, however, has led to a consensus opinion that cells of the myeloid lineage are one site of carriage of HCMV in vivo and that in myeloid dendritic cell (DC) progenitors the viral genome is carried latently in the absence of virus lytic gene expression. In contrast, differentiation of these cells to a mature DC phenotype is linked with reactivation of infectious virus resulting from differentiation-dependent chromatin remodelling of the viral major immediate-early promoter. Thus there is a crucial link between the differentiation of myeloid cells and transcriptional reactivation of latent virus which is likely to play a key role in viral pathogenesis.
Collapse
Affiliation(s)
- John Sinclair
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK.
| |
Collapse
|
122
|
Identification of the DNA binding element of the human ZNF300 protein. Cell Mol Biol Lett 2008; 13:391-403. [PMID: 18350257 PMCID: PMC6275660 DOI: 10.2478/s11658-008-0005-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Accepted: 12/04/2007] [Indexed: 11/20/2022] Open
Abstract
The human ZNF300 gene is a member of the KRAB/C2H2 zinc finger gene family, the members of which are known to be involved in various developmental and pathological processes. Here, we show that the ZNF300 gene encodes a 68-kDa nuclear protein that binds DNA in a sequence-specific manner. The ZNF300 DNA binding site, C(t/a)GGGGG(c/g)G, was defined via a random oligonucleotide selection assay, and the DNA binding site was further confirmed by electrophoretic mobility shift assays. A potential ZNF300 binding site was found in the promoter region of the human IL-2Rβ gene. The results of electrophoretic mobility shift assays indicated that ZNF300 bound to the ZNF300 binding site in the IL-2Rβ promoter in vitro. Transient co-transfection assays showed that ZNF300 could activate the IL-2Rβ promoter, and that the activation was abrogated by the mutation of residues in the ZNF300 binding site. Identifying the DNA binding site and characterizing the transcriptional regulation property of ZNF300 would provide critical insights into its potential as a transcriptional regulator.
Collapse
|
123
|
The LIM protein AJUBA recruits protein arginine methyltransferase 5 to mediate SNAIL-dependent transcriptional repression. Mol Cell Biol 2008; 28:3198-207. [PMID: 18347060 DOI: 10.1128/mcb.01435-07] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The SNAIL transcription factor contains C-terminal tandem zinc finger motifs and an N-terminal SNAG repression domain. The members of the SNAIL family have recently emerged as major contributors to the processes of development and metastasis via the regulation of epithelial-mesenchymal transition events during embryonic development and tumor progression. However, the mechanisms by which SNAIL represses gene expression are largely undefined. Previously we demonstrated that the AJUBA family of LIM proteins function as corepressors for SNAIL and, as such, may serve as a platform for the assembly of chromatin-modifying factors. Here, we describe the identification of the protein arginine methyltransferase 5 (PRMT5) as an effector recruited to SNAIL through an interaction with AJUBA that functions to repress the SNAIL target gene, E-cadherin. PRMT5 binds to the non-LIM region of AJUBA and is translocated into the nucleus in a SNAIL- and AJUBA-dependent manner. The depletion of PRMT5 in p19 cells stimulates E-cadherin expression, and the SNAIL, AJUBA, and PRMT5 ternary complex can be found at the proximal promoter region of the E-cadherin gene, concomitant with increased arginine methylation of histones at the locus. Together, these data suggest that PRMT5 is an effector of SNAIL-dependent gene repression.
Collapse
|
124
|
Zarebski A, Velu CS, Baktula AM, Bourdeau T, Horman SR, Basu S, Bertolone SJ, Horwitz M, Hildeman DA, Trent JO, Grimes HL. Mutations in growth factor independent-1 associated with human neutropenia block murine granulopoiesis through colony stimulating factor-1. Immunity 2008; 28:370-80. [PMID: 18328744 DOI: 10.1016/j.immuni.2007.12.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Revised: 11/15/2007] [Accepted: 12/21/2007] [Indexed: 12/14/2022]
Abstract
Severe congenital neutropenia (SCN) is characterized by a deficiency of mature neutrophils, leading to recurrent bacterial and fungal infections. Although mutations in Elastase-2, neutrophil (ELA2) predominate in human SCN, mutation of Ela2 in mice does not recapitulate SCN. The growth factor independent-1 (GFI1) transcription factor regulates ELA2. Mutations in GFI1 are associated with human SCN, and genetic deletion of Gfi1 results in murine neutropenia. We examined whether human SCN-associated GFI1N382S mutant proteins are causal in SCN and found that GFI1 functions as a rate-limiting granulopoietic molecular switch. The N382S mutation inhibited GFI1 DNA binding and resulted in a dominant-negative block to murine granulopoiesis. Moreover, Gfi1N382S selectively derepressed the monopoietic cytokine CSF1 and its receptor. Gfi1N382S-expressing Csf1-/- cells formed neutrophils. These results reveal a common transcriptional program that underlies both human and murine myelopoiesis, and that is central to the pathogenesis of SCN associated with mutations in GFI1. This shared transcriptional pathway may provide new avenues for understanding SCN caused by mutations in other genes and for clinical intervention into human neutropenias.
Collapse
Affiliation(s)
- Adrian Zarebski
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
The zinc finger protein and transcriptional repressor Gfi1 as a regulator of the innate immune response. Immunobiology 2008; 213:341-52. [PMID: 18406379 DOI: 10.1016/j.imbio.2007.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 10/25/2007] [Accepted: 11/02/2007] [Indexed: 12/13/2022]
Abstract
Gfi1 is a transcriptional repressor with a molecular weight between 47 and 55kDa. The protein has six C-terminal C(2)H(2)-type zinc finger domains and a characteristic stretch of 20 amino acids, called the SNAG-domain, at its N-terminus. Expression of Gfi1 ranges from the hematopoietic and lymphoid system to sensory epithelia, lung and parts of the CNS. Gene knockout studies revealed that Gfi1 is essential for the development of granulocytes and plays a role in macrophage-dependent cytokine production, indicating that this protein shares the responsibility for different lines of defense against pathogens. Strikingly, Gfi1-deficient mice are highly sensitive to both endotoxin and bacterial infections and die rapidly after an experimental application of endotoxin or induction of infection with symptoms of septic shock. This sensitivity is mediated by an overproduction of tumor necrosis factor (TNF) and other inflammatory cytokines. The lung could be identified as the principal organ in which the accelerated inflammatory reactions take place in challenged Gfi1-deficient mice. Several lines of experimental evidence support a role of Gfi1 as a regulator of the Toll-like receptor (TLR) pathways, and, in general, as an essential modulator preventing an overshooting of the inflammatory response.
Collapse
|
126
|
Abstract
Primary infection of healthy individuals with human cytomegalovirus (HCMV) is usually asymptomatic and results in the establishment of a lifelong latent infection of the host. Although no overt HCMV disease is observed in healthy carriers, due to effective immune control, severe clinical symptoms associated with HCMV reactivation are observed in immunocompromised transplant patients and HIV sufferers. Work from a number of laboratories has identified the myeloid lineage as one important site for HCMV latency and reactivation and thus has been the subject of extensive study. Attempts to elucidate the mechanisms controlling viral latency have shown that cellular transcription factors and histone proteins influence HCMV gene expression profoundly and that the type of cellular environment virus encounters upon infection may have a critical role in determining a lytic or latent infection and subsequent reactivation from latency. Furthermore, the identification of a number of viral gene products expressed during latent infection suggests a more active role for HCMV during latency. Defining the role of these viral proteins in latently infected cells will be important for our full understanding of HCMV latency and reactivation in vivo.
Collapse
|
127
|
ZHAO DX, TENG XC, DING ZC, HUANG ZX. Expression and Purification of ZNF191(243–368) in Three Expression Systems. CHINESE J CHEM 2007. [DOI: 10.1002/cjoc.200790321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
128
|
Ayyanathan K, Peng H, Hou Z, Fredericks WJ, Goyal RK, Langer EM, Longmore GD, Rauscher FJ. The Ajuba LIM domain protein is a corepressor for SNAG domain mediated repression and participates in nucleocytoplasmic Shuttling. Cancer Res 2007; 67:9097-106. [PMID: 17909014 DOI: 10.1158/0008-5472.can-07-2987] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The SNAG repression domain is comprised of a highly conserved 21-amino acid sequence, is named for its presence in the Snail/growth factor independence-1 class of zinc finger transcription factors, and is present in a variety of proto-oncogenic transcription factors and developmental regulators. The prototype SNAG domain containing oncogene, growth factor independence-1, is responsible for the development of T cell thymomas. The SNAIL proteins also encode the SNAG domain and play key roles in epithelial mesenchymal differentiation events during development and metastasis. Significantly, these oncogenic functions require a functional SNAG domain. The molecular mechanisms of SNAG domain-mediated transcriptional repression are largely unknown. Using a yeast two-hybrid strategy, we identified Ajuba, a multiple LIM domain protein that can function as a corepressor for the SNAG domain. Ajuba interacts with the SNAG domain in vitro and in vivo, colocalizes with it, and enhances SNAG-mediated transcriptional repression. Ajuba shuttles between the cytoplasm and the nucleus and may form a novel intracellular signaling system. Using an integrated reporter gene combined with chromatin immunoprecipitation, we observed rapid, SNAG-dependent assembly of a multiprotein complex that included Ajuba, SNAG, and histone modifications consistent with the repressed state. Thus, SNAG domain proteins may bind Ajuba, trapping it in the nucleus where it functions as an adapter or molecular scaffold for the assembly of macromolecular repression complexes at target promoters.
Collapse
|
129
|
Xie B, Charlton-Perkins M, McDonald E, Gebelein B, Cook T. Senseless functions as a molecular switch for color photoreceptor differentiation in Drosophila. Development 2007; 134:4243-53. [PMID: 17978002 DOI: 10.1242/dev.012781] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A major question in development is how different specialized cell types arise from a common progenitor. In the adult Drosophila compound eye, color discrimination is achieved by UV-, blue- and green-sensitive photoreceptors (PRs). These different PR subsets arise from neuronal precursors called R7 and R8 cells. Recent studies have demonstrated that R7-based UV-sensitive PRs require the repression of R8-based blue/green-sensitive PR characteristics to properly develop. This repression is mediated by the transcription factor Prospero (Pros). Here, we report that Senseless (Sens), a Drosophila ortholog of the vertebrate Gfi1 transcription factor, plays an opposing role to Pros by both negatively regulating R7-based features and positively enforcing R8-based features during terminal differentiation. In addition, we demonstrate that Pros and Sens function together with the transcription factor Orthodenticle (Otd) to oppositely regulate R7 and R8 PR Rhodopsin gene expression in vitro. These data show that sens, previously shown to be essential for neuronal specification, also controls differentiation of specific neuronal subtypes in the retina. Interestingly, Pros has recently been shown to function as a tumor suppressor, whereas Gfi1 is a well-characterized oncogene. Thus, we propose that sens/pros antagonism is important for regulating many biological processes.
Collapse
Affiliation(s)
- Baotong Xie
- Department of Pediatric Ophthalmology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
130
|
|
131
|
Bharadwaj AS, Agrawal DK. Transcription factors in the control of dendritic cell life cycle. Immunol Res 2007; 37:79-96. [PMID: 17496348 DOI: 10.1007/bf02686091] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that guard all parts of the body. They have the unique ability to prime T cells and generate primary immune responses. Their journey beginning with the development from precursor cells and ending with their death is controlled by a group of transcription factors. Some of the transcription factors like PU.1 are involved in more than one stage of DC life. Other transcription factors including Ikaros and JAK3 are involved in the development of more than one cell type. For a long time, the cellular and molecular mechanisms underlying the development, differentiation, maturation, and other stages of DC life were not well understood. However, in recent years novel information has been published by many researchers to better understand the molecular mechanisms of the development and function of DCs in immunological diseases such as asthma, cancer, autoimmunity, and transplantation. This review will discuss the various transcription factors and signaling pathways involved in each stage of the life cycle of DCs.
Collapse
Affiliation(s)
- Arpita S Bharadwaj
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | |
Collapse
|
132
|
Saban R, Simpson C, Davis CA, Dozmorov I, Maier J, Fowler B, Ihnat MA, Hurst RE, Wershil BK, Saban MR. Transcription factor network downstream of protease activated receptors (PARs) modulating mouse bladder inflammation. BMC Immunol 2007; 8:17. [PMID: 17705868 PMCID: PMC2000913 DOI: 10.1186/1471-2172-8-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Accepted: 08/17/2007] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND All four PARs are present in the urinary bladder, and their expression is altered during inflammation. In order to search for therapeutic targets other than the receptors themselves, we set forth to determine TFs downstream of PAR activation in the C57BL/6 urinary bladders. METHODS For this purpose, we used a protein/DNA combo array containing 345 different TF consensus sequences. Next, the TF selected was validated by EMSA and IHC. As mast cells seem to play a fundamental role in bladder inflammation, we determined whether c-kit receptor deficient (Kit w/Kit w-v) mice have an abrogated response to PAR stimulation. Finally, TFEB antibody was used for CHIP/Q-PCR assay and revealed up-regulation of genes known to be downstream of TFEB. RESULTS TFEB, a member of the MiTF family of basic helix-loop-helix leucine zipper, was the only TF commonly up-regulated by all PAR-APs. IHC results confirm a correlation between inflammation and TFEB expression in C57BL/6 mice. In contrast, Kit w/Kit w-v mice did not exhibit inflammation in response to PAR activation. EMSA results confirmed the increased TFEB binding activity in C57BL/6 but not in Kit w/Kit w-v mice. CONCLUSION This is the first report describing the increased expression of TFEB in bladder inflammation in response to PAR activation. As TFEB belongs to a family of TFs essential for mast cell survival, our findings suggest that this molecule may influence the participation of mast cells in PAR-mediated inflammation and that targeting TFEB/MiTF activity may be a novel approach for the treatment of bladder inflammatory disorders.
Collapse
Affiliation(s)
- Ricardo Saban
- Department of Physiology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Cindy Simpson
- Department of Physiology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Carole A Davis
- Department of Physiology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Igor Dozmorov
- Oklahoma Medical Research Foundation (OMRF), Imaging Core Facility, Oklahoma City, Oklahoma 73104, USA
| | - Julie Maier
- Oklahoma Medical Research Foundation (OMRF), Arthritis and Immunology Research Program, Microarray/Euk. Genomics Core Facility, Oklahoma City, Oklahoma 73104. USA
| | - Ben Fowler
- Oklahoma Medical Research Foundation (OMRF), Arthritis and Immunology Research Program, Microarray/Euk. Genomics Core Facility, Oklahoma City, Oklahoma 73104. USA
| | - Michael A Ihnat
- Department of Cell Biology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Robert E Hurst
- Department of Urology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Barry K Wershil
- Albert Einstein College of Medicine Division of Pediatric GI and Nutrition The Children's Hospital at Montefiore Bronx, NY 10467, USA
| | - Marcia R Saban
- Department of Physiology, The University Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
133
|
Duan Z, Person RE, Lee HH, Huang S, Donadieu J, Badolato R, Grimes HL, Papayannopoulou T, Horwitz MS. Epigenetic regulation of protein-coding and microRNA genes by the Gfi1-interacting tumor suppressor PRDM5. Mol Cell Biol 2007; 27:6889-902. [PMID: 17636019 PMCID: PMC2099216 DOI: 10.1128/mcb.00762-07] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gfi1 transcriptionally governs hematopoiesis, and its mutations produce neutropenia. In an effort to identify Gfi1-interacting proteins and also to generate new candidate genes causing neutropenia, we performed a yeast two-hybrid screen with Gfi1. Among other Gfi1-interacting proteins, we identified a previously uncharacterized member of the PR domain-containing family of tumor suppressors, PRDM5. PRDM5 has 16 zinc fingers, and we show that it acts as a sequence-specific, DNA binding transcription factor that targets hematopoiesis-associated protein-coding and microRNA genes, including many that are also targets of Gfi1. PRDM5 epigenetically regulates transcription similarly to Gfi1: it recruits the histone methyltransferase G9a and class I histone deacetylases to its target gene promoters and demonstrates repressor activity on synthetic reporters; on endogenous target genes, however, it functions as an activator, in addition to a repressor. Interestingly, genes that PRDM5 activates, as opposed to those it represses, are also targets of Gfi1, suggesting a competitive mechanism through which two repressors could cooperate in order to become transcriptional activators. In neutropenic patients, we identified PRDM5 protein sequence variants perturbing transcriptional function, suggesting a potentially important role in hematopoiesis.
Collapse
Affiliation(s)
- Zhijun Duan
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Jiang L, Tang D, Wang K, Zhang H, Yuan C, Duan D, Xiao X. Functional analysis of a novel KRAB/C2H2 zinc finger protein Mipu1. Biochem Biophys Res Commun 2007; 356:829-35. [PMID: 17397802 DOI: 10.1016/j.bbrc.2007.02.138] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 02/19/2007] [Indexed: 11/23/2022]
Abstract
A novel rat gene, Mipu1, encodes a 608 amino acid protein with an amino-terminal KRAB domain and 14 carboxyl-terminal C2H2 zinc finger motifs. Mipu1 is localized to the nucleus through its KRAB domain or the linker adjacent to its zinc finger region. Using the GST-Mipu1 bound to glutathione-Sepharose beads, a consensus putative DNA binding site (5'-TGTCTTATCGAA-3') was extracted from a random oligonucleotide library. EMSA and target detection assay showed that the probe containing the putative site can bind to purified GST-Mipu1 fusion protein. The oligonucleotide containing the putative site was inserted into the pGL3-promotor vector to produce a reporter construct. The expression of reporter gene was repressed by overexpression of Mipu1 in a dose-dependent manner. Mutation analysis of the consensus sequence indicated that the repression mediated by Mipu1 is sequence-dependent. These results suggest that Mipu1 is a nuclear protein, which functions as a transcriptional repressor.
Collapse
Affiliation(s)
- Lei Jiang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, PR China
| | | | | | | | | | | | | |
Collapse
|
135
|
Abstract
The induction of Bcl-x(L) is critical for the survival of late proerythroblasts. The erythroid-specific transcriptional network that regulates Bcl-x(L) expression in erythropoiesis remains unclear. The activation of the central erythropoietic transcriptional factor, GATA-1, leads to the early, transient induction of a transcription repressor, Gfi-1B, followed by the late induction of Bcl-x(L) during erythroid maturation in G1ER cells. Chromatin immunoprecipitation assays demonstrated that a constant level of GATA-1 binds to the Bcl-x promoter throughout the entire induction period, while Gfi-1B is transiently associated with the promoter in the early phase. The sustained expression of Gfi-1B abolished GATA-1-induced Bcl-x(L) expression. Here, we present evidence that GATA-1 binds to the noncanonical GATT motif of the Bcl-x promoter for trans-activation. Gfi-1B expressed at increased levels is recruited to the Bcl-x promoter through its association with GATA-1, suppressing Bcl-x(L) transcription. Therefore, the down-regulation of Gfi-1B in the late phase of erythroid maturation is necessary for Bcl-x(L) induction. Furthermore, we show that the inhibition of Bcr-Abl kinase by treatment with imatinib caused the up-regulation of Gfi-1B in K562 cells, where Gfi-1B also cooperated with GATA-1 to repress Bcl-x(L) transcription. Gfi-1B knockdown by RNA interference diminished imatinib-induced apoptosis, while the overexpression of Gfi-1B sensitized K562 cells to arsenic-induced death. These findings illuminate the role of Gfi-1B in GATA-1-mediated transcription in the survival aspect of erythroid cells.
Collapse
Affiliation(s)
- Yuan-Yeh Kuo
- Institute of Biochemistry and Molecular Biology, National Taiwan University, College of Medicine, Taipei, Taiwan, Republic of China
| | | |
Collapse
|
136
|
Rathinam C, Klein C. Transcriptional repressor Gfi1 integrates cytokine-receptor signals controlling B-cell differentiation. PLoS One 2007; 2:e306. [PMID: 17375192 PMCID: PMC1810430 DOI: 10.1371/journal.pone.0000306] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Accepted: 02/20/2007] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic stem cell differentiation is specified by cytokines and transcription factors, but the mechanisms controlling instructive and permissive signalling networks are poorly understood. We provide evidence that CLP1-dependent IL7-receptor mediated B cell differentiation is critically controlled by the transcriptional repressor Gfi1. Gfi1-deficient progenitor B cells show global defects in IL7Rα-dependent signal cascades. Consequently, IL7-dependent trophic, proliferative and differentiation-inducing responses of progenitor B cells are perturbed. Gfi1 directly regulates expression levels of IL7Rα and indirectly controls STAT5 signalling via expression of SOCS3. Thus, Gfi1 selectively specifies IL7-dependent development of B cells from CLP1 progenitors, providing clues to the transcriptional networks integrating cytokine signals and lymphoid differentiation.
Collapse
Affiliation(s)
- Chozhavendan Rathinam
- Department of Pediatric Hematology/Oncology, Hannover Medical School, Hannover, Germany
| | - Christoph Klein
- Department of Pediatric Hematology/Oncology, Hannover Medical School, Hannover, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
137
|
Rosenbauer F, Tenen DG. Transcription factors in myeloid development: balancing differentiation with transformation. Nat Rev Immunol 2007; 7:105-17. [PMID: 17259967 DOI: 10.1038/nri2024] [Citation(s) in RCA: 446] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In recent years, great progress has been made in elucidating the progenitor-cell hierarchy of the myeloid lineage. Transcription factors have been shown to be key determinants in the orchestration of myeloid identity and differentiation fates. Most transcription factors show cell-lineage-restricted and stage-restricted expression patterns, indicating the requirement for tight regulation of their activities. Moreover, if dysregulated or mutated, these transcription factors cause the differentiation block observed in many myeloid leukaemias. Consequently, therapies designed to restore defective transcription factor functions are an attractive option in the treatment of myeloid and other human cancers.
Collapse
Affiliation(s)
- Frank Rosenbauer
- Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13092 Berlin, Germany.
| | | |
Collapse
|
138
|
Nakazawa Y, Suzuki M, Manabe N, Yamada T, Kihara-Negishi F, Sakurai T, Tenen DG, Iwama A, Mochizuki M, Oikawa T. Cooperative interaction between ETS1 and GFI1 transcription factors in the repression of Bax gene expression. Oncogene 2007; 26:3541-50. [PMID: 17213822 DOI: 10.1038/sj.onc.1210140] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The proto-oncoproteins ETS1 and growth factor independent-1 (GFI1) are implicated in cell growth and differentiation in various types of cells, and their deregulated expression is involved in malignant transformation. Here, we report that ETS1 and GFI1 interact and affect gene expression through their cross-talk. Co-immunoprecipitation analyses and glutathione-S-transferase pull-down assays revealed that ETS1 bound directly to GFI1 via its Ets domain, and GFI1 bound to ETS1 via its zinc-finger domain. Luciferase (Luc) assays using artificial reporters showed that GFI1 repressed ETS1-mediated transcriptional activation and ETS1 repressed GFI1-mediated transcriptional activation, in a dose-dependent manner. However, in the Bax promoter where the Ets- and Gfi-binding sites (EBS and GBS) are adjacent, ETS1 and GFI1 cooperatively reduced activation. Site-directed mutagenesis on the EBS and GBS of the Bax promoter showed that both binding sites were necessary for full repression. Chromatin immunoprecipitation analyses confirmed that an ETS1-GFI1 complex formed on the Bax promoter even when either EBS or GBS was mutated. Introduction of small interfering RNA against ETS1 and/or GFI1 enhanced endogenous Bax gene expression. Our results suggest that the interaction between ETS1 and GFI1 facilitates their binding to specific sites on the Bax promoter and represses Bax expression in vivo.
Collapse
Affiliation(s)
- Y Nakazawa
- Department of Cell Genetics, Sasaki Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Uchimoto T, Itoga S, Nezu M, Sunaga M, Tomonaga T, Nomura F. Role of the Genetic Polymorphisms in the 5'-Flanking Region for Transcriptional Regulation of the Human CYP2E1 Gene. Alcohol Clin Exp Res 2007; 31:S36-42. [PMID: 17331164 DOI: 10.1111/j.1530-0277.2006.00284.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The human cytochrome P-4502E1 (CYP2E1) has been known to be that are associated with alcohol-related diseases and various cancers. Difference in susceptibility in enzyme induction of the CYP2E1 by environmental factors such as ethanol may be associated with the risk of these diseases. In the 5'-flanking region of the human CYP2E1 gene, there were tandem repeat polymorphism (CYP2E1*1C*,1D) and RsaI polymorphism (CYP2E1*5A*,5B), which have been reported to be related to these diseases. Thus, we focused relationship between these polymorphisms and transcriptional regulation. The aim of this study was to determine the role of these polymorphisms for the transcriptional regulation of the human CYP2E1 gene in general transcription and during enzyme induction. METHODS To determine the role of these polymorphisms, various constructs of the 5'-flanking region of the human CYP2E1 gene were prepared and transfected into HeLa cells and HepG2 cells treated or not with pyrazine, a well-known CYP2E1 inducer. RESULTS The transcriptional activity of CYP2E1*1D was higher than that of CYP2E1*1C in HeLa cells. The -1,306 base pairs (bp) construct (-1,306 to +9) increased transcriptional activity compared with the 2.5 kb full-length construct. The suppressive effect of transcriptional activity by deletion of the region including CYP2E1*1C was greater than that by CYP2E1*1D in HeLa cells. The -580 bp construct (-580 to +9) decreased transcriptional activity compared with the -1,306 bp construct. The -1,306 bp construct also showed a similar tendency during pyrazine treatment. CONCLUSIONS The region including the tandem repeat polymorphism was suggested to regulate transcription suppressively. Besides, it was speculated that the transcriptional activity of CYP2E1*1D was higher than that of CYP2E1*1C because transcriptional suppression of CYP2E1*1D was weaker than that of CYP2E1*1C. Also, it was confirmed that the region including RsaI polymorphism was associated with the promotion of transcription.
Collapse
Affiliation(s)
- Takayuki Uchimoto
- Department of Molecular Diagnosis (F8), Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
140
|
Dahl R, Iyer SR, Owens KS, Cuylear DD, Simon MC. The transcriptional repressor GFI-1 antagonizes PU.1 activity through protein-protein interaction. J Biol Chem 2006; 282:6473-83. [PMID: 17197705 PMCID: PMC3218793 DOI: 10.1074/jbc.m607613200] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Mice lacking the zinc finger transcriptional repressor protein GFI-1 are neutropenic. These mice generate abnormal immature myeloid cells exhibiting characteristics of both macrophages and granulocytes. Furthermore, Gfi-1(-/-) mice are highly susceptible to bacterial infection. Interestingly, Gfi-1(-/-) myeloid cells overexpress target genes of the PU.1 transcription factor such as the macrophage colony-stimulating factor receptor and PU.1 itself. We therefore determined whether GFI-1 modulates the transcriptional activity of PU.1. Our data demonstrate that GFI-1 physically interacts with PU.1, repressing PU.1-dependent transcription. This repression is functionally significant, as GFI-1 blocked PU.1-induced macrophage differentiation of a multipotential hematopoietic progenitor cell line. Retroviral expression of GFI-1 in primary murine hematopoietic progenitors increased granulocyte differentiation at the expense of macrophage differentiation. We interbred Gfi-1(+/-) and PU.1(+/-) mice and observed that heterozygosity at the PU.1 locus partially rescued the Gfi-1(-/-) mixed myeloid lineage phenotype, but failed to restore granulocyte differentiation. Our data demonstrate that GFI-1 represses PU.1 activity and that lack of this repression in Gfi-1(-/-) myeloid cells contributes to the observed mixed lineage phenotype.
Collapse
Affiliation(s)
- Richard Dahl
- Department of Internal Medicine, Health Sciences Center, University of New Mexico, Albuquerque, New Mexico 87131, USA.
| | | | | | | | | |
Collapse
|
141
|
Vassen L, Okayama T, Möröy T. Gfi1b:green fluorescent protein knock-in mice reveal a dynamic expression pattern of Gfi1b during hematopoiesis that is largely complementary to Gfi1. Blood 2006; 109:2356-64. [PMID: 17095621 DOI: 10.1182/blood-2006-06-030031] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Gfi1b and Gfi1 are 37- and 55-kDa transcriptional repressors that share common features such as a 20-amino acid (aa) N-terminal SNAG domain, a nonconserved intermediary domain, and 6 highly conserved C-terminal zinc fingers. Both gene loci are under autoregulatory and cross-regulatory feedback control. We have generated a reporter mouse strain by inserting the cDNA for green fluorescent protein (GFP) into the Gfi1b gene locus which allowed us to follow Gfi1b expression during hematopoiesis and lymphopoiesis by measuring green fluorescence. We found highly dynamic expression patterns of Gfi1b in erythroid cells, megakaryocytes, and their progenitor cells (MEPS) where Gfi1 is not detected. Vice versa, Gfi1b could not be found in granulocytes, activated macrophages, or their granulomonocytic precursors (GMPs) or in mature naive or activated lymphocytes where Gfi1 is expressed, suggesting a complementary regulation of both loci during hematopoiesis. However, Gfi1b was found to be up-regulated in early stages of B-cell and in a subset of early T-cell development, where Gfi1 is also present, suggesting that cross-regulation of both loci exists but is cell-type specific.
Collapse
Affiliation(s)
- Lothar Vassen
- Institut für Zellbiologie (Tumorforschung), Universitätsklinikum Essen, Germany
| | | | | |
Collapse
|
142
|
Marteijn JAF, van der Meer LT, Van Emst L, de Witte T, Jansen JH, van der Reijden BA. Diminished proteasomal degradation results in accumulation of Gfi1 protein in monocytes. Blood 2006; 109:100-8. [PMID: 16888099 DOI: 10.1182/blood-2006-02-003590] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Gfi1 is a transcriptional repressor essential during myeloid differentiation. Gfi1−/− mice exhibit a block in myeloid differentiation resulting in the accumulation of an immature myelo-monocytic cell population and the complete absence of mature neutrophils. Even though mRNA levels of Gfi1 appear to be very low in monocytes, Gfi1 might play a role in the monocytic lineage as Gfi1−/− mice exhibit diminished monocyte-derived dendritic cells and disturbed cytokine production by macrophages in response to LPS. We show here that Gfi1 protein levels are mainly regulated by the ubiquitin-proteasome system. Upon forced monocytic differentiation of U937 cells, Gfi1 mRNA levels dropped but protein levels increased due to diminished proteasomal turnover. Similarly, Gfi1 mRNA levels are low in primary monocytes whereas the protein is clearly detectable. Conversely, Gfi1 mRNA levels are high in granulocytes but the protein is swiftly degraded by the proteasome in these cells. Chromatin immunoprecipitation experiments showed that Gfi1 binds to the promoter of several granulocyte-specific genes in primary monocytes, including C/EBPα, neutrophil elastase, and Gfi1 itself. The binding of the repressor Gfi1 to these promoters correlated with low expression of these genes in monocytes compared with granulocytes. Our data fit a model in which Gfi1 protein levels are induced in primary monocytes, due to diminished proteasomal degradation, to repress genes that play a role in granulocytic differentiation.
Collapse
Affiliation(s)
- Jurgen A F Marteijn
- Central Hematology Laboratory, Radboud University Nijmegen Medical Centre for Molecular Life Sciences (NCMLS), PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
143
|
Abstract
Human cytomegalovirus (HCMV) persists as a subclinical, lifelong infection in the normal human host, maintained at least in part by its carriage in the absence of detectable infectious virus – the hallmark of latent infection. Reactivation from latency in immunocompromised individuals, in contrast, often results in serious disease. Latency and reactivation are defining characteristics of the herpesviruses and key to understanding their biology. However, the precise cellular sites in which HCMV is carried and the mechanisms regulating its latency and reactivation during natural infection remain poorly understood. This review will detail our current knowledge of where HCMV is carried in healthy individuals, which viral genes are expressed upon carriage of the virus and what effect this has on cellular gene expression. It will also address the accumulating evidence suggesting that reactivation of HCMV from latency appears to be linked intrinsically to the differentiation status of the myeloid cell, and how the cellular mechanisms that normally control host gene expression play a critical role in the differential regulation of viral gene expression during latency and reactivation.
Collapse
Affiliation(s)
- John Sinclair
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Patrick Sissons
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
144
|
Grassmé H, Jin J, Wilker B, von Kürthy G, Wick W, Weller M, Möröy T, Gulbins E. Regulation of pulmonary Pseudomonas aeruginosa infection by the transcriptional repressor Gfi1. Cell Microbiol 2006; 8:1096-105. [PMID: 16819963 DOI: 10.1111/j.1462-5822.2006.00690.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pulmonary Pseudomonas aeruginosa infections are characterized by the release of proinflammatory mediators, focal induction of apoptosis in respiratory epithelial cells and internalization of the bacteria. Here, we demonstrate that the transcriptional regulator Gfi1 is critically involved in the regulation of proinflammatory cytokine release and the induction of apoptosis in respiratory epithelial cells and macrophages upon P. aeruginosa infection. Gfi1-deficient mice responded to a pulmonary P. aeruginosa infection with uncontrolled pulmonary release of interleukin (IL)-1 and tumour necrosis factor (TNF)-alpha, sepsis and death, which were delayed by injection of IL-1- and TNF-alpha-neutralizing antibodies. The uncontrolled release of cytokines seems to be caused by a failure of Gfi1-deficient respiratory epithelial cells in large to small bronchi and macrophages to respond to P. aeruginosa infection with an induction of apoptosis. Pharmacological inhibition of apoptosis in wild-type mice by intravenous injection of the broad-spectrum caspase inhibitor zVAD-cmk mimicked the phenotype of Gfi1-deficient mice and resulted in a profound sensitization of mice to P. aeruginosa, an increased release of cytokines, sepsis and death of the animals. Thus, Gfi1 controls apoptosis of respiratory epithelial cells and macrophages upon infection with P. aeruginosa. Inhibition of apoptosis by Gfi1 deficiency or caspase blockers sensitizes mice to P. aeruginosa infections, suggesting that apoptosis functions as a novel defence mechanisms in the regulation of the local innate immune response.
Collapse
Affiliation(s)
- Heike Grassmé
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Kazanjian A, Gross EA, Grimes HL. The growth factor independence-1 transcription factor: new functions and new insights. Crit Rev Oncol Hematol 2006; 59:85-97. [PMID: 16716599 PMCID: PMC2830489 DOI: 10.1016/j.critrevonc.2006.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Revised: 01/30/2006] [Accepted: 02/01/2006] [Indexed: 12/21/2022] Open
Abstract
The growth factor independence-1 (Gfi1) transcription factor is required for proper development of neuroendocrine cells, sensory neurons, and blood. Patients with mutations in Gfi1 exhibit severe congenital neutropenia (SCN) or non-immune chronic idiopathic neutropenia of adults. Gfi1 was initially described as an oncoprotein that mediates tumor progression in a mouse model of leukemia; however, recent data suggest that Gfi1 may act as either an oncogene or an anti-proliferative tumor suppressor gene depending on the cell type. Here we review the latest literature on Gfi1, and emphasize its role in the hematopoietic, sensory and neuroendocrine systems.
Collapse
Affiliation(s)
- Avedis Kazanjian
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Eleanore A. Gross
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - H. Leighton Grimes
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Surgery, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Corresponding author. Present address: Cincinnati Children’s Hospital Medical Center, Division of Immunobiology – MLC 7038, 3333 Burnet Avenue, Room 5564, Cincinnati, OH 45229-3039, USA. Tel.: +1 513 636 6089; fax: +1 513 636 5355.
| |
Collapse
|
146
|
Acar M, Jafar-Nejad H, Giagtzoglou N, Yallampalli S, David G, He Y, Delidakis C, Bellen HJ. Senseless physically interacts with proneural proteins and functions as a transcriptional co-activator. Development 2006; 133:1979-89. [PMID: 16624856 DOI: 10.1242/dev.02372] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The zinc-finger transcription factor Senseless is co-expressed with basic helix-loop-helix (bHLH) proneural proteins in Drosophila sensory organ precursors and is required for their normal development. High levels of Senseless synergize with bHLH proteins and upregulate target gene expression,whereas low levels of Senseless act as a repressor in vivo. However, the molecular mechanism for this dual role is unknown. Here, we show that Senseless binds bHLH proneural proteins via its core zinc fingers and is recruited by proneural proteins to their target enhancers to function as a co-activator. Some point mutations in the Senseless zinc-finger region abolish its DNA-binding ability but partially spare the ability of Senseless to synergize with proneural proteins and to induce sensory organ formation in vivo. Therefore, we propose that the structural basis for the switch between the repressor and co-activator functions of Senseless is the ability of its core zinc fingers to interact physically with both DNA and bHLH proneural proteins. As Senseless zinc fingers are ∼90% identical to the corresponding zinc fingers of its vertebrate homologue Gfi1, which is thought to cooperate with bHLH proteins in several contexts, the Senseless/bHLH interaction might be evolutionarily conserved.
Collapse
Affiliation(s)
- Melih Acar
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Vassen L, Fiolka K, Möröy T. Gfi1b alters histone methylation at target gene promoters and sites of gamma-satellite containing heterochromatin. EMBO J 2006; 25:2409-19. [PMID: 16688220 PMCID: PMC1478184 DOI: 10.1038/sj.emboj.7601124] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2005] [Accepted: 04/11/2006] [Indexed: 01/30/2023] Open
Abstract
Gfi1b is a 37 kDa nuclear protein with six C2H2 zinc-finger domains that can silence transcription upon binding to specific target gene promoters. Here we show by using a chromatin immunoprecipitation and cloning protocol that Gfi1b also binds to gamma-satellite sequences that mainly occur in pericentric heterochromatin. Immuno-FISH experiments demonstrated that Gfi1b is localized at foci of pericentric heterochromatin identified by DAPI staining. Elevated levels of Gfi1b correlated with increased histone H3 lysine 9 dimethylation at sites neighboring gamma-satellite sequences but also at Gfi1b target gene promoters. In Gfi1b-deficient cells, however, a decrease of histone H3 lysine 9 trimethylation and a loss of heterochromatic structures was observed. Strikingly, we found that Gfi1b binds to both SUV39H1 and G9A histone methyl transferases, which provides a direct link between histone methylation and Gfi1b at heterochromatic and euchromatic sites. We propose that Gfi1b functions in heterochromatin formation and silencing of euchromatic transcription by recruiting histone methyl transferases to either gamma-satellite sequences or specific target gene promoters.
Collapse
Affiliation(s)
- Lothar Vassen
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Essen, Germany
| | - Katharina Fiolka
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Essen, Germany
| | - Tarik Möröy
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Essen, Germany
- Present address: Institut de recherches cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, Québec, Canada H2W 1R7. E-mail:
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Virchowstrasse 173, 45122 Essen, Germany. Tel.: +49 201 723 3380; Fax: +49 201 723 5904; E-mail:
| |
Collapse
|
148
|
Gao J, Scott JG. Role of the transcriptional repressor mdGfi-1 in CYP6D1v1-mediated insecticide resistance in the house fly, Musca domestica. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2006; 36:387-95. [PMID: 16651185 DOI: 10.1016/j.ibmb.2006.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 02/01/2006] [Accepted: 02/02/2006] [Indexed: 05/08/2023]
Abstract
Gfi-1 is a C(2)H(2)-type zinc finger protein that is a transcriptional repressor in vertebrates and has been implicated in control of CYP6D1 expression in house flies (Musca domestica). A 15 bp insert, which disrupts a putative mdGfi-1 binding site in the CYP6D1v1 promoter has been implicated as a cause of increased expression of CYP6D1, and thus insecticide resistance. Using electrophoretic mobility shift assays we demonstrate that the CYP6D1 promoter from susceptible strains binds mdGfi-1. The 15 bp insert that interrupts the mdGfi-1-binding site in insecticide-resistant strains reduces the amount of mdGfi-1 binding by 9- to 20-fold, consistent with the role of mdGfi-1 in resistance. Partial sequences of mdGfi-1 (spanning the first intron) from individual houseflies from 11 different strains revealed the presence of 23 alleles. There was no consistent difference in the mdGfi-1 alleles between susceptible and CYP6D1-mediated insecticide-resistant strains, indicating that mdGfi-1 alleles were not likely involved in resistance. Polymorphisms were used to map mdGfi-1 to autosome 1. Quantitative real time PCR (qRT-PCR) revealed Gfi-1 expression was higher in the thorax compared to the head and abdomen, and varied between life stages and between strains. However, similar levels of mdGfi-1 were detected in susceptible and resistant adults suggesting that altered levels of mdGfi-1 were not likely a cause of insecticide resistance. The significance of these results to understanding insecticide resistance is discussed.
Collapse
Affiliation(s)
- Jianwei Gao
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, NY 14853-0901, USA
| | | |
Collapse
|
149
|
Blondheim NR, Levy YS, Ben-Zur T, Burshtein A, Cherlow T, Kan I, Barzilai R, Bahat-Stromza M, Barhum Y, Bulvik S, Melamed E, Offen D. Human Mesenchymal Stem Cells Express Neural Genes, Suggesting a Neural Predisposition. Stem Cells Dev 2006; 15:141-64. [PMID: 16646662 DOI: 10.1089/scd.2006.15.141] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Because of their unique attributes of plasticity and accessibility, bone marrow-derived mesenchymal stem cells (MSCs) may find use for therapy of neurodegenerative disorders. Our previous studies of adult human MSCs demonstrated that these cells express an extensive assortment of neural genes at a low but clearly detectable level. Here, we report expression of 12 neural genes, 8 genes related to the neuro-dopaminergic system, and 11 transcription factors with neural significance by human MSCs. Our results suggest that, as opposed to cells that do not express neural genes, human MSCs are predisposed to differentiate to neuronal and glial lineages, given the proper conditions. Our findings add a new dimension in which to view adult stem cell plasticity, and may explain the relative ease with which MSCs, transplanted into the central nervous system (CNS) differentiate to a variety of functional neural cell types. Our results further promote the possibility that adult human MSCs are promising candidates for cell-based therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Netta R Blondheim
- Laboratory of Neurosciences, Felsenstein Medical Research Center and Department of Neurology, Rabin Medical Center, Beilinson Campus Tel Aviv University, Sackler School of Medicine, Petah-Tikva 49100, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Jafar-Nejad H, Tien AC, Acar M, Bellen HJ. Senseless and Daughterless confer neuronal identity to epithelial cells in the Drosophila wing margin. Development 2006; 133:1683-92. [PMID: 16554363 DOI: 10.1242/dev.02338] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The basic helix-loop-helix (bHLH) proneural proteins Achaete and Scute cooperate with the class I bHLH protein Daughterless to specify the precursors of most sensory bristles in Drosophila. However, the mechanosensory bristles at the Drosophila wing margin have been reported to be unaffected by mutations that remove Achaete and Scute function. Indeed, the proneural gene(s) for these organs is not known. Here, we show that the zinc-finger transcription factor Senseless, together with Daughterless, plays the proneural role for the wing margin mechanosensory precursors, whereas Achaete and Scute are required for the survival of the mechanosensory neuron and support cells in these lineages. We provide evidence that Senseless and Daughterless physically interact and synergize in vivo and in transcription assays. Gain-of-function studies indicate that Senseless and Daughterless are sufficient to generate thoracic sensory organs (SOs) in the absence of achaete-scute gene complex function. However, analysis of senseless loss-of-function clones in the thorax implicates Senseless not in the primary SO precursor (pI) selection, but in the specification of pI progeny. Therefore, although Senseless and bHLH proneural proteins are employed during the development of all Drosophila bristles, they play fundamentally different roles in different subtypes of these organs. Our data indicate that transcription factors other than bHLH proteins can also perform the proneural function in the Drosophila peripheral nervous system.
Collapse
Affiliation(s)
- Hamed Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | |
Collapse
|