901
|
Yang TTC, Xiong Q, Graef IA, Crabtree GR, Chow CW. Recruitment of the extracellular signal-regulated kinase/ribosomal S6 kinase signaling pathway to the NFATc4 transcription activation complex. Mol Cell Biol 2005; 25:907-20. [PMID: 15657420 PMCID: PMC544015 DOI: 10.1128/mcb.25.3.907-920.2005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integration of protein kinases into transcription activation complexes influences the magnitude of gene expression. The nuclear factor of activated T cells (NFAT) group of proteins are critical transcription factors that direct gene expression in immune and nonimmune cells. A balance of phosphotransferase activity is necessary for optimal NFAT activation. Activation of NFAT requires dephosphorylation by the calcium-mediated calcineurin phosphatase to promote NFAT nuclear accumulation, and the Ras-activated extracellular signal-regulated kinase (ERK) mitogen-activated protein (MAP) kinase, which targets NFAT partners, to potentiate transcription. Whether protein kinases operate on NFAT and contribute positively to transcription activation is not clear. Here, we coupled DNA affinity isolation with in-gel kinase assays to avidly pull down the activated NFAT and identify its associated protein kinases. We demonstrate that p90 ribosomal S6 kinase (RSK) is recruited to the NFAT-DNA transcription complex upon activation. The formation of RSK-NFATc4-DNA transcription complex is also apparent upon adipogenesis. Bound RSK phosphorylates Ser(676) and potentiates NFATc4 DNA binding by escalating NFAT-DNA association. Ser(676) is also targeted by the ERK MAP kinase, which interacts with NFAT at a distinct region than RSK. Thus, integration of the ERK/RSK signaling pathway provides a mechanism to modulate NFATc4 transcription activity.
Collapse
Affiliation(s)
- Teddy T C Yang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
902
|
Marafioti T, Marafiot T, Pozzobon M, Hansmann ML, Ventura R, Pileri SA, Roberton H, Gesk S, Gaulard P, Barth TFE, Du MQ, Leoncini L, Möller P, Natkunam Y, Siebert R, Mason DY. The NFATc1 transcription factor is widely expressed in white cells and translocates from the cytoplasm to the nucleus in a subset of human lymphomas. Br J Haematol 2005; 128:333-42. [PMID: 15667535 DOI: 10.1111/j.1365-2141.2004.05313.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Stimulation of lymphoid cells via their surface receptors triggers signalling pathways that terminate in the nucleus, where they induce alterations in gene transcription. Nuclear factor of activated T cells (NFAT) transcription factors, involved in a major Ca2+-dependent signalling pathway, normally reside in the cytoplasm but re-locate to the nucleus when activation of the pathway (e.g. following ligation of antigen receptors) leads to their dephosphorylation. This study found that one member of the NFAT family (NFATc1/NFAT2) can be detected in routine biopsy samples, where it is seen in essentially all lymphoid cells, but is absent from the great majority of non-haematopoietic cells. An immunohistological evaluation of NFATc1 in almost 300 lymphomas showed that most neoplastic lymphoid cells also express NFATc1 as a cytoplasmic constituent, although it is absent in classical Hodgkin's disease and plasma cell proliferations. Of particular interest was the finding that NFATc1 was relocated to the nucleus in a minority of lymphoid neoplasms (usually diffuse large B-cell lymphomas or Burkitt lymphoma), presumably reflecting activation of the NFAT pathway. It would be of interest to correlate this feature with patterns of gene expression and also with prognosis, since it may identify a subset of human lymphoma that is distinct in its molecular and clinical features.
Collapse
Affiliation(s)
- Teresa Marafioti
- Leukaemia Research Fund Immunodiagnostics Unit, Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
903
|
Ortega-Pérez I, Cano E, Were F, Villar M, Vázquez J, Redondo JM. c-Jun N-terminal kinase (JNK) positively regulates NFATc2 transactivation through phosphorylation within the N-terminal regulatory domain. J Biol Chem 2005; 280:20867-78. [PMID: 15743762 DOI: 10.1074/jbc.m501898200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The nuclear factor of activated T cells (NFAT) family of transcription factors regulates the transcription of cytokine genes and other genes involved in the regulation and function of the immune system. NFAT activity is regulated by the phosphatase calcineurin, which binds and dephosphorylates the NFAT N-terminal regulatory domain, a critical step required for nuclear translocation and transcriptional activity. Here we show that the mitogen-activated protein kinase (MAPK) JNK activates NFATc2-dependent transcription. Mass spectrometry revealed that JNK phosphorylates at least six residues within the NFATc2 regulatory domain in vitro. Transfection of cells with a chimeric construct encoding the GAL-4 DNA binding domain linked to wild-type NFATc2 showed that JNK stimulates the NFATc2 transactivation domain in activated Jurkat T lymphocytes, an effect that is inhibited by dominant-negative versions of JNK. Likewise, the mutation of the phosphorylation sites identified revealed that Thr(116) and Ser(170) are critical for the transactivation of NFATc2 by JNK. In addition, clustered mutation of the SP-conserved motifs of NFATc2 showed that SP1 and SP2, but not SP3, are also important for the inducible transactivation of NFATc2. Furthermore, mass spectrometry analysis of NFATc2-transfected cells indicated that the activation of the JNK pathway results in the in vivo phosphorylation of Thr(116). Our results indicate that, unlike other NFAT members, the transcriptional activity of NFATc2 is up-regulated by JNK. JNK-mediated phosphorylation of NFATs thus appears to play a differential physiological role among NFAT family members.
Collapse
Affiliation(s)
- Inmaculada Ortega-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Ronda de Poniente 5, Tres Cantos, Madrid 28760, Spain
| | | | | | | | | | | |
Collapse
|
904
|
Duque J, Fresno M, Iñiguez MA. Expression and Function of the Nuclear Factor of Activated T Cells in Colon Carcinoma Cells. J Biol Chem 2005; 280:8686-93. [PMID: 15632146 DOI: 10.1074/jbc.m413076200] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence shows a crucial role of the Ca2+/ calcineurin-mediated activation of the nuclear factor of activated T cells (NFAT) in the regulation of a variety of processes in nonimmune cells. Here we provide evidence that NFATc1 and NFATc2 are expressed in human colon carcinoma cell lines. These proteins are translocated from the cytoplasm to the nucleus upon treatment with a combination of phorbol 12-myristate 13-acetate plus the calcium ionophore A23187. Subsequent to translocation to the nucleus, NFATc1 and NFATc2 were able to bind to a NFAT response element in the DNA, regulating transcriptional activation of genes containing a NFAT-responsive element such as cyclooxygenase-2 (COX-2). COX-2 expression and prostaglandin E2 (PGE2) production were induced upon pharmacological stimuli leading to NFAT activation and blunted by inhibition of calcineurin phosphatase with cyclosporin A or tacrolimus (FK506). Expression of NFAT wild type protein or the active catalytic subunit of calcineurin transactivates COX-2 promoter activity, whereas a dominant negative mutant of NFAT inhibited COX-2 induction in colon carcinoma cell lines. Furthermore, mutation or deletion of NFAT binding sites in the human COX-2 promoter greatly diminished its induction by phorbol 12-myristate 13-acetate/calcium ionophore A23187. These findings demonstrate the presence and activation of NFAT in human colon carcinoma cells, with important implications in the regulation of genes involved in the transformed phenotype as COX-2.
Collapse
Affiliation(s)
- Javier Duque
- Centro de Biología Molecular Severo Ochoa, Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | | | | |
Collapse
|
905
|
Rodríguez A, Martínez-Martínez S, López-Maderuelo MD, Ortega-Pérez I, Redondo JM. The Linker Region Joining the Catalytic and the Regulatory Domains of CnA Is Essential for Binding to NFAT. J Biol Chem 2005; 280:9980-4. [PMID: 15671033 DOI: 10.1074/jbc.c400401200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calcineurin (CN) is an important regulator of developmental processes and in adults controls the immune response through its regulation of nuclear factor of activated T cells (NFAT). The physical interaction between CN and NFATs is an essential step in the activation of NFAT-dependent genes by calcium signals. Using deletional and substitutional analyses, we have identified a 13-amino acid region within CN that is essential for the interaction with NFAT and with two other CN-binding proteins, AKAP79 and Cabin-1. The interaction of CN with these proteins is selectively disrupted by substitution of specific amino acid residues within this region, indicating that NFAT and other CN-interacting proteins bind differentially to CN. This selectivity suggests that the region identified in CN could be a potential molecular target for immunosuppressive and other therapeutic interventions in diseases involving the CN/NFAT pathway.
Collapse
Affiliation(s)
- Antonio Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Ronda de Poniente 5, Tres Cantos, Madrid 28760, Spain
| | | | | | | | | |
Collapse
|
906
|
Kimmel AR, Firtel RA. Breaking symmetries: regulation of Dictyostelium development through chemoattractant and morphogen signal-response. Curr Opin Genet Dev 2005; 14:540-9. [PMID: 15380246 DOI: 10.1016/j.gde.2004.08.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dictyostelium discoideum grow unicellularly, but develop as multicellular organisms. At two stages of development, their underlying symmetrical pattern of cellular organization becomes disrupted. During the formation of the multicellular aggregate, individual non-polarized cells re-organize their cytoskeletal structures to sequester specific intracellular signaling elements for activation by and directed movement within chemoattractant gradients. Subsequently, response to secreted morphogens directs undifferentiated populations to adopt different cell fates. Using a combination of cellular, biochemical and molecular approaches, workers have now begun to understand the mechanisms that permit Dictyostelium (and other chemotactic cells) to move directionally in shallow chemoattractant gradients and the transcriptional regulatory pathways that polarize cell-fate choice and initiate pattern formation.
Collapse
Affiliation(s)
- Alan R Kimmel
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, Maryland 20892-8028, USA.
| | | |
Collapse
|
907
|
Johnson EN, Appelbaum ER, Carpenter DC, Cox RF, Disa J, Foley JJ, Ghosh SK, Naselsky DP, Pullen MA, Sarau HM, Scheff SR, Steplewski KM, Zaks-Zilberman M, Aiyar N. Neuromedin U elicits cytokine release in murine Th2-type T cell clone D10.G4.1. THE JOURNAL OF IMMUNOLOGY 2005; 173:7230-8. [PMID: 15585845 DOI: 10.4049/jimmunol.173.12.7230] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuromedin U (NmU), originally isolated from porcine spinal cord and later from other species, is a novel peptide that potently contracts smooth muscle. NmU interacts with two G protein-coupled receptors designated as NmU-1R and NmU-2R. This study demonstrates a potential proinflammatory role for NmU. In a mouse Th2 cell line (D10.G4.1), a single class of high affinity saturable binding sites for (125)I-labeled NmU (K(D) 364 pM and B(max) 1114 fmol/mg protein) was identified, and mRNA encoding NmU-1R, but not NmU-2R, was present. Competition binding analysis revealed equipotent, high affinity binding of NmU isopeptides to membranes prepared from D10.G4.1 cells. Exposure of these cells to NmU isopeptides resulted in an increase in intracellular Ca(2+) concentration (EC(50) 4.8 nM for human NmU). In addition, NmU also significantly increased the synthesis and release of cytokines including IL-4, IL-5, IL-6, IL-10, and IL-13. Studies using pharmacological inhibitors indicated that maximal NmU-evoked cytokine release required functional phospholipase C, calcineurin, MEK, and PI3K pathways. These data suggest a role for NmU in inflammation by stimulating cytokine production by T cells.
Collapse
Affiliation(s)
- Eric N Johnson
- Department of High Throughput Biology, GlaxoSmithKline, Research Triangle Park, NC 27709, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
908
|
Komarova SV, Pereverzev A, Shum JW, Sims SM, Dixon SJ. Convergent signaling by acidosis and receptor activator of NF-kappaB ligand (RANKL) on the calcium/calcineurin/NFAT pathway in osteoclasts. Proc Natl Acad Sci U S A 2005; 102:2643-8. [PMID: 15695591 PMCID: PMC548977 DOI: 10.1073/pnas.0406874102] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Systemic acidosis has detrimental effects on the skeleton, and local acidosis coincides with bone destruction in inflammatory and metastatic diseases. Acidification dramatically enhances osteoclastic resorption, although the underlying mechanism has remained elusive. We investigated the effect of acidosis on the osteoclastogenic transcription factor NFATc1, which upon dephosphorylation translocates from the cytoplasm to nuclei. Lowering extracellular pH dramatically increased accumulation of NFATc1 in nuclei of rat and rabbit osteoclasts to levels comparable with those induced by the proresorptive cytokine receptor activator of NF-kappaB ligand (RANKL). Activation of NFATc1 by RANKL was mediated by means of prolonged stimulation of the Ca2+/calmodulin-dependent protein phosphatase, calcineurin. In contrast, NFATc1 activation by acidosis involved stimulation of calcineurin and suppression of NFATc1 inactivation. Acidosis, like RANKL, induced transient elevation of cytosolic free Ca2+ concentration ([Ca2+]i), which persisted in Ca2+-free media and was abolished by inhibition of phospholipase C or depletion of intracellular Ca2+ stores. Real-time-PCR of osteoclast-like cells generated from RAW 264.7 cells revealed high levels of expression of ovarian cancer G protein-coupled receptor 1, which links extracellular acidification to elevation of [Ca2+]i. In addition, the calcineurin inhibitor cyclosporin A suppressed the stimulatory effect of acidification on resorption, implicating NFAT in mediating the actions of acidosis on osteoclast activity. In summary, acidification and RANKL induce signals in osteoclasts that converge on the Ca2+/calcineurin/NFAT pathway. Acidosis acts directly on osteoclasts to activate NFATc1 and stimulate resorption.
Collapse
Affiliation(s)
- Svetlana V Komarova
- Canadian Institutes of Health Research Group in Skeletal Development and Remodeling, Department of Physiology and Pharmacology, Faculty of Medicine and Dentistry, University of Western Ontario, London, ON, Canada N6A 5C1
| | | | | | | | | |
Collapse
|
909
|
Abstract
As thousands of new genes are identified in genomics efforts, the rush is on to learn something about the functional roles of the proteins encoded by those genes. Clues to protein functions, activation states and protein-protein interactions have been revealed in focused studies of protein localization. With technical breakthroughs such as GFP protein tagging and recombinase cloning systems, large-scale screens of protein localization are now being undertaken.
Collapse
Affiliation(s)
- Nancy A O'Rourke
- Alliance for Cell Signaling, Microscopy Lab, Stanford University School of Medicine, 975 California Ave, Palo Alto, CA 94304, USA
| | | | | |
Collapse
|
910
|
Takayanagi H. Mechanistic insight into osteoclast differentiation in osteoimmunology. J Mol Med (Berl) 2005; 83:170-9. [PMID: 15776286 DOI: 10.1007/s00109-004-0612-6] [Citation(s) in RCA: 305] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Accepted: 10/22/2004] [Indexed: 02/06/2023]
Abstract
Recently a close relationship between the immune and skeletal systems or the interdisciplinary field called osteoimmunology has attracted much attention due to the observations that bone destruction is caused by an abnormal activation of the immune system in rheumatoid arthritis, and that mice lacking immunomodulatory molecules often exhibit an unexpected bone phenotype. Osteoclasts are cells of monocyte/macrophage origin that degrade the bone matrix. They are among the key players in the control of bone metabolism in health and disease. Receptor activator of NF-kappaB ligand (RANKL), a tumor necrosis factor (TNF) family cytokine, induces the differentiation of osteoclasts in the presence of macrophage-colony stimulating factor. RANKL activates TRAF6, c-Fos, and calcium signaling pathways, all of which are indispensable for the induction and activation of nuclear factor of activated T cells (NFAT) c1, the master transcription factor for osteoclastogenesis. The autoamplification of NFATc1 gene results in the efficient induction of osteoclast-specific genes. An AP-1 transcription factor complex containing c-Fos plays a crucial role in these processes, although results in conditional knockout mice show that Jun family members have a redundant role. The immunoreceptor tyrosine-based activation motif (ITAM) is an important signaling component for a number of receptors in the immune system including T-cell, B-cell, NK-cell, and Fc receptors, but its contribution to the skeletal system remains unclarified. In search for the calcium-mobilizing mechanism during osteoclastogenesis we determined that multiple immunoglobulinlike receptors associated with ITAM-harboring adaptors, Fc receptor common gamma chain (FcRgamma), and DNAX-activating protein (DAP) 12, are essential for osteoclastogenesis. In osteoclast precursor cells FcRgamma-associated receptors include osteoclast-associated receptor and paired immunoglobulinlike receptor A, while triggering receptor expressed in myeloid cells 2 and signal-regulatory protein beta1 preferentially associate with DAP12. In cooperation with RANKL these receptors activate phospholipase Cgamma and calcium signaling essential for the induction of NFATc1 through ITAM phosphorylation. Thus we have established the importance of the ITAM-mediated costimulatory signals in RANKL-induced osteoclast differentiation, which is analogous to the role of costimulatory signals in the immune system. Here we summarize recent advances in the study of signaling mechanism of osteoclast differentiation in the context of osteoimmunology.
Collapse
Affiliation(s)
- Hiroshi Takayanagi
- Department of Cellular Physiological Chemistry, COE Program for Frontier Research on Molecular Destruction and Reconstruction of Tooth and Bone, Graduate School, Tokyo Medical and Dental University, Japan.
| |
Collapse
|
911
|
Affiliation(s)
- Monte M Winslow
- Immunology Program, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
912
|
Badou A, Basavappa S, Desai R, Peng YQ, Matza D, Mehal WZ, Kaczmarek LK, Boulpaep EL, Flavell RA. Requirement of voltage-gated calcium channel beta4 subunit for T lymphocyte functions. Science 2005; 307:117-21. [PMID: 15637280 DOI: 10.1126/science.1100582] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Calcium is known to play vital roles in diverse physiological processes, and it is known that voltage-gated calcium channels (Cav) mediate calcium influx in excitable cells. However, no consensus exists on the molecular identity of the calcium channels present in nonexcitable cells such as T lymphocytes. Here, we demonstrate that T lymphocytes express both regulatory beta4 and poreforming Cav1 alpha1 subunits of Cav channels. Cav beta4-mutant T lymphocytes fail to acquire normal functions and display impairment in the calcium response, activation of the transcription factor NFAT, and cytokine production. Although Cav1 channels of lymphocytes retain their voltage dependency, T cell receptor stimulation dramatically increases channel opening, providing a new mechanism for calcium entry in lymphocytes.
Collapse
Affiliation(s)
- Abdallah Badou
- Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
913
|
Jayanthi S, Deng X, Ladenheim B, McCoy MT, Cluster A, Cai NS, Cadet JL. Calcineurin/NFAT-induced up-regulation of the Fas ligand/Fas death pathway is involved in methamphetamine-induced neuronal apoptosis. Proc Natl Acad Sci U S A 2005; 102:868-73. [PMID: 15644446 PMCID: PMC545515 DOI: 10.1073/pnas.0404990102] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Methamphetamine [METH ("speed")] is an abused psychostimulant that can cause psychotic, cognitive, and psychomotor impairment in humans. These signs and symptoms are thought to be related to dysfunctions in basal ganglionic structures of the brain. To identify possible molecular bases for these clinical manifestations, we first used cDNA microarray technology to measure METH-induced transcriptional responses in the striatum of rats treated with an apoptosis-inducing dose of the drug. METH injection resulted in increased expression of members of the Jun, Egr, and Nur77 subfamilies of transcription factors (TFs), changes that were confirmed by quantitative PCR. Because pathways linked to these factors are involved in the up-regulation of Fas ligand (FasL), FasL mRNA was quantified and found to be increased. Immunohistochemical studies also revealed METH-induced increased FasL protein expression in striatal GABAergic neurons that express enkephalin. Moreover, there were METH-mediated increases in calcineurin, as well as shuttling of nuclear factor of activated T cells (NFAT)c3 and NFATc4 from the cytosol to the nucleus of METH-treated rats, mechanisms also known to be involved in FasL regulation. Furthermore, METH induced cleavage of caspase-3 in FasL- and Fas-containing neurons. Finally, the METH-induced changes in the FasL-Fas death pathway were attenuated by pretreatment with the dopamine D1 receptor antagonist, SCH23390, which also caused attenuation of METH-induced apoptosis. These observations indicate that METH causes some of its neurodegenerative effects, in part, via stimulation of the Fas-mediated cell death pathway consequent to FasL up-regulation mediated by activation of multiple TFs.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Department of Health and Human Services, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
914
|
Benedito AB, Lehtinen M, Massol R, Lopes UG, Kirchhausen T, Rao A, Bonni A. The Transcription Factor NFAT3 Mediates Neuronal Survival. J Biol Chem 2005; 280:2818-25. [PMID: 15537643 DOI: 10.1074/jbc.m408741200] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal apoptosis is critical for normal development of the mammalian nervous system and also contributes to the pathogenesis of ischemic and degenerative diseases of the brain. Apoptosis of neurons is tightly regulated by extrinsic signals including growth factors and neuronal activity, but the intracellular mechanisms by which these signals promote neuronal survival are incompletely understood. We report that the transcription factor NFAT3 plays a critical role in mediating survival of granule neurons of the developing cerebellum. NFAT3 accumulated in the nucleus of primary granule neurons under survival conditions of serum growth factors and neuronal activity that was elicited by depolarization with high K(+). In contrast, deprivation of serum and K(+), which leads to neuronal apoptosis, triggered NFAT3 nuclear export. Treatment of granule neurons with Li(+), an inhibitor of the NFAT export kinase GSK3, prevented the nuclear export of NFAT3 and increased granule cell survival even under pro-apoptotic conditions. Thus, the nuclear localization of NFAT3 correlated tightly with granule neuron survival. Consistent with a pro-survival function for NFAT3, genetic knockdown of NFAT3 by RNA interference in primary granule neurons led to increased apoptosis even in neurons cultured under survival conditions. Conversely, expression of a constitutively active form of NFAT protected neurons against apoptosis induced by serum withdrawal and low K(+). Taken together, these results reveal an essential function for NFAT3-mediated transcription in neuronal survival that may play important roles in the developing and mature brain.
Collapse
Affiliation(s)
- Alessandra B Benedito
- Center for Blood Research, Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
915
|
Abstract
NF-kappaB and c-Fos are transcription factors that are activated in immune cells and in most other cell types following stimulation by a variety of factors, including cytokines, growth factors, and hormones. They regulate the expression of a large number of genes, and both are activated in osteoclast precursors after RANKL, IL-1, or TNF bind to their respective receptors. However, of these cytokines, only RANKL is required for the induction of osteoclast formation in vivo. Nevertheless, it is likely that IL-1, TNF, and other cytokines participate in the upregulation of osteoclast formation seen in a variety of conditions that affect the skeleton in which cytokine production is increased, including estrogen deficiency and inflammatory bone diseases. In this review, the RANKL/ OPG/RANK system and roles for NF-kappaB and c-Fos in osteoclasts are reviewed along with our current understanding of how this system may be disrupted in common bone diseases, such as postmenopausal osteoporosis, inflammatory arthritis, and Paget's disease.
Collapse
Affiliation(s)
- Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY 14642, USA.
| | | | | | | | | | | |
Collapse
|
916
|
Tsika G, Ji J, Tsika R. Sp3 proteins negatively regulate beta myosin heavy chain gene expression during skeletal muscle inactivity. Mol Cell Biol 2004; 24:10777-91. [PMID: 15572681 PMCID: PMC533985 DOI: 10.1128/mcb.24.24.10777-10791.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In adult skeletal muscle, beta myosin heavy chain (betaMyHC) gene expression is primarily restricted to slow type I fibers; however, its expression is down-regulated in response to muscle inactivity. Little is known about the signaling pathways and transcription factors that mediate this important functional response. This study demonstrates that increased binding of Sp3 to GC-rich elements in the betaMyHC promoter is a critical event in down-regulation of betaMyHC gene expression under non-weight-bearing conditions. Conversely, binding of Sp3 to these elements decreased while Sp1 binding increased with nuclear extracts from plantaris muscle exposed to mechanical overload, a stimulus that increases betaMyHC gene expression. In addition, these experiments revealed the existence of an Sp4-DNA binding complex when using adult skeletal muscle nuclear extract was used but not when nuclear extracts from cultured myotubes were used. Sp3 proteins are competitive inhibitors of Sp1-mediated betaMyHC reporter gene transactivation in both Drosophila SL-2 and mouse C2C12 myotubes. Sp4 is a weak activator of betaMyHC gene expression in SL-2 cells, which lack endogenous Sp1 activity, but does not activate betaMyHC gene expression in C2C12 myotubes, which have high levels of Sp1. These results suggest that competitive binding of Sp family proteins regulate betaMyHC gene transcription in response to altered neuromuscular activity.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- Blotting, Western
- Cell Nucleus/metabolism
- Cells, Cultured
- DNA Footprinting
- DNA-Binding Proteins/metabolism
- Electrophoretic Mobility Shift Assay
- GC Rich Sequence
- Gene Expression Regulation
- Genes, Regulator
- Genes, Reporter
- Luciferases/metabolism
- Mice
- Molecular Sequence Data
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Slow-Twitch/metabolism
- Muscle, Skeletal/physiology
- Mutagenesis, Site-Directed
- Myosin Heavy Chains/chemistry
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Promoter Regions, Genetic
- Rats
- Sequence Homology, Amino Acid
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Sp3 Transcription Factor
- Transcription Factors/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Gretchen Tsika
- Department of Biochemistry, School of Medicine, University of Missouri-Columbia, Biochemistry E102 Vet Med Bldg., 1600 Rollins Road, Columbia, MO 65211, USA.
| | | | | |
Collapse
|
917
|
Salazar C, Höfer T. Activation of the transcription factor NFAT1: concerted or modular regulation? FEBS Lett 2004; 579:621-6. [PMID: 15670818 DOI: 10.1016/j.febslet.2004.12.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2004] [Revised: 12/08/2004] [Accepted: 12/13/2004] [Indexed: 10/26/2022]
Abstract
The transcription factor NFAT1 is activated through dephosphorylation of multiple serine residues, contained within the SRR1 and SP motifs. The phosphorylation status of these motifs regulates the subcellular localisation of NFAT1 via a conformational switch. Here, we discuss two molecular mechanisms for NFAT1 activation that resemble network-oriented approaches. In the modular mechanism, import and export are regulated separately by the SRR1 and SP motifs, respectively, whereas in the concerted model all residues jointly control both processes. Using simulations of a computational model, we show that both mechanisms may be compatible with recent experimental data on the import and export kinetics of NFAT1.
Collapse
Affiliation(s)
- Carlos Salazar
- Theoretische Biophysik, Institut für Biologie, Humboldt-Universität zu Berlin, Invalidenstrasse 42, 10115 Berlin, Germany.
| | | |
Collapse
|
918
|
Grassberger M, Steinhoff M, Schneider D, Luger TA. Pimecrolimus - an anti-inflammatory drug targeting the skin. Exp Dermatol 2004; 13:721-30. [PMID: 15560755 DOI: 10.1111/j.0906-6705.2004.00269.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pimecrolimus is the most recent member of calcineurin inhibitors available for the therapy for inflammatory skin diseases. It targets T-cells and mast cells and inhibits the production and release of cytokines and other inflammatory mediators, as well as the expression of signals essential for the activation of inflammatory T-lymphocytes. Pimecrolimus has a cell-selective mode of action. In contrast to corticosteroids, it does not affect, e.g., Langerhans'cells/dendritic cells (LC/DC), as demonstrated in vitro with human monocyte-derived DC and in vivo with epidermal LC in mice, nor human primary fibroblasts. As shown in vitro with human skin and by comparison of clinical pharmacokinetic data from patients with atopic dermatitis, pimecrolimus permeates less through skin than tacrolimus and much less than corticosteroids. It, thus, has a lower potential for transcutaneous resorption after topical administration, resulting in a lower risk of systemic effects. Pimecrolimus has high anti-inflammatory activity in animal models of skin inflammation, including a model reflecting neurogenic inflammation, but a more favourable balance of anti-inflammatory vs. immunosuppressive activity than tacrolimus. Pimecrolimus does not affect sensitization in a murine model of allergic contact dermatitis and has a lower potency in various models of immunosuppression after systemic administration, compared to tacrolimus. In conclusion, the results of preclinical studies show that pimecrolimus has a selective pharmacological profile, suited for effective and safe treatment for inflammatory skin diseases.
Collapse
Affiliation(s)
- M Grassberger
- Novartis Institute for Biomedical Research, Vienna, Austria.
| | | | | | | |
Collapse
|
919
|
Zhu LL, Zaidi S, Moonga BS, Troen BR, Sun L. RANK-L induces the expression of NFATc1, but not of NFκB subunits during osteoclast formation. Biochem Biophys Res Commun 2004; 326:131-5. [PMID: 15567162 DOI: 10.1016/j.bbrc.2004.10.212] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Indexed: 11/18/2022]
Abstract
We report the program of gene expression during osteoclast formation from RAW264.7 cell precursors in response to RANK-ligand (RANK-L) using a combination of quantitative real time PCR and Affymetrix gene chip assays. We found that genes obligatory to osteoclast formation and function, namely tartrate-resistant acid phosphatase, cathepsin K, beta3 integrin, and calcitonin receptors, were up-regulated by RANK-L markedly by up to approximately 2000-fold. In contrast, we found a cluster of genes that were significantly down-regulated: these included interleukin-18, insulin-like growth factor-1, interleukin-6 receptor, and cathepsins B, C, and L. These results from real time PCR were broadly concordant with those obtained from Affymetrix. We also explored the expression of the transcription factors of the NFAT and NFkappaB family at days 3 and 5 of culture. Whereas NFATc1 expression was increased significantly at days 3 and 5 following RANK-L exposure, there were no significant increases in the expression of NFkappaB subunits, namely p65, p50, c-Rel, IkappaBalpha, and IkappaBbeta. There were also no significant differences in transcription modulator expression between days 3 and 5, except for c-Rel and NFATc4, which were both decreased significantly at day 5. The studies suggest RANK-L regulates the expression only of NFATc1, while it signals through both NFATc1 and NFkappaB.
Collapse
Affiliation(s)
- Ling-Ling Zhu
- Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
920
|
Role of intracellular calcium signaling in the pathophysiology and pharmacotherapy of bipolar disorder: current status. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.cnr.2004.09.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
921
|
Quintana A, Griesemer D, Schwarz EC, Hoth M. Calcium-dependent activation of T-lymphocytes. Pflugers Arch 2004; 450:1-12. [PMID: 15806400 DOI: 10.1007/s00424-004-1364-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Accepted: 10/18/2004] [Indexed: 12/11/2022]
Abstract
Activation of T-lymphocytes requires stimulation of T-cell receptors (TCR) and co-stimulatory signals. Among different signalling cascades, TCR engagement induces Ca(2+) entry through plasma membrane Ca(2+) channels, which is an indispensable step for T-cells to expand clonally and to acquire effector functions. The Ca(2+) channels are activated by depletion of Ca(2+) stores and are called Ca(2+) release-activated Ca(2+) (CRAC) channels. Ca(2+) influx through CRAC channels is also controlled, directly or indirectly, by K(+) channels, Ca(2+)-ATPases, mitochondria, endoplasmic reticulum and Ca(2+) buffers. We review the functional implications of these transporters, organelles and buffers and develop a model of Ca(2+) signal generation that depends mainly on their relative mutual localization. This model offers the possibility of controlling amplitude and kinetics of Ca(2+) signals in T-cells. Decoding of various Ca(2+) signals allows differential activation of the transcription factor families nuclear factor of activated T-cells (NFAT), nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1). Variation of amplitude and kinetics of Ca(2+) signals thus is an important mechanism for modulating the specificity of T-cell responses.
Collapse
Affiliation(s)
- Ariel Quintana
- Institut für Physiologie, Universität des Saarlandes, Gebäude 58, 66421 Homburg/Saar, Germany
| | | | | | | |
Collapse
|
922
|
Wilkins BJ, Molkentin JD. Calcium-calcineurin signaling in the regulation of cardiac hypertrophy. Biochem Biophys Res Commun 2004; 322:1178-91. [PMID: 15336966 DOI: 10.1016/j.bbrc.2004.07.121] [Citation(s) in RCA: 348] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Indexed: 12/21/2022]
Abstract
Cardiac hypertrophy is a leading predicator of progressive heart disease that often leads to heart failure and a loss of cardiac contractile performance associated with profound alterations in intracellular calcium handling. Recent investigation has centered on identifying the molecular signaling pathways that regulate cardiac myocyte hypertrophy, as well as the mechanisms whereby alterations in calcium handling are associated with progressive heart failure. One potential focal regulator of cardiomyocyte hypertrophy that also responds to altered calcium handling is the calmodulin-activated serine/threonine protein phosphatase calcineurin (PP2B). Once activated by increases in calcium, calcineurin mediates the hypertrophic response through its downstream transcriptional effector nuclear factor of activated T cells (NFAT), which is directly dephosphorylated by calcineurin resulting in nuclear translocation. While previous studies have convincingly demonstrated the sufficiency of calcineurin to mediate cardiac hypertrophy and progressive heart failure, its necessity remains an area of ongoing investigation. Here we weigh an increasing body of literature that suggests a causal link between calcineurin signaling and the cardiac hypertrophic response and heart failure through the use of pharmacologic inhibitors (cyclosporine A and FK506) and genetic approaches. We will also discuss the manner in which calcineurin-NFAT signaling is negatively regulated in the heart through a diverse array of kinases and inhibitory proteins. Finally, we will discuss emerging theories as to the mechanisms whereby alterations in intracellular calcium handling might stimulate calcineurin within the context of a contractile cell continually experiencing calcium flux.
Collapse
Affiliation(s)
- Benjamin J Wilkins
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | |
Collapse
|
923
|
Frey N, Barrientos T, Shelton JM, Frank D, Rütten H, Gehring D, Kuhn C, Lutz M, Rothermel B, Bassel-Duby R, Richardson JA, Katus HA, Hill JA, Olson EN. Mice lacking calsarcin-1 are sensitized to calcineurin signaling and show accelerated cardiomyopathy in response to pathological biomechanical stress. Nat Med 2004; 10:1336-43. [PMID: 15543153 DOI: 10.1038/nm1132] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Accepted: 09/27/2004] [Indexed: 11/08/2022]
Abstract
Signaling by the calcium-dependent phosphatase calcineurin profoundly influences the growth and gene expression of cardiac and skeletal muscle. Calcineurin binds to calsarcins, a family of muscle-specific proteins of the sarcomeric Z-disc, a focal point in the pathogenesis of human cardiomyopathies. We show that calsarcin-1 negatively modulates the functions of calcineurin, such that calcineurin signaling was enhanced in striated muscles of mice that do not express calsarcin-1. As a consequence of inappropriate calcineurin activation, mice with a null mutation in calsarcin-1 showed an excess of slow skeletal muscle fibers. The absence of calsarcin-1 also activated a hypertrophic gene program, despite the absence of hypertrophy, and enhanced the cardiac growth response to pressure overload. In contrast, cardiac adaptation to other hypertrophic stimuli, such as chronic catecholamine stimulation or exercise, was not affected. These findings show important roles for calsarcins as modulators of calcineurin signaling and the transmission of a specific subset of stress signals leading to cardiac remodeling in vivo.
Collapse
Affiliation(s)
- Norbert Frey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9148, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
924
|
Abdel-Razzak Z, Garlatti M, Aggerbeck M, Barouki R. Determination of interleukin-4-responsive region in the human cytochrome P450 2E1 gene promoter. Biochem Pharmacol 2004; 68:1371-81. [PMID: 15345327 DOI: 10.1016/j.bcp.2004.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Accepted: 06/04/2004] [Indexed: 11/26/2022]
Abstract
Cytochrome P450 2E1 (CYP2E1) gene expression is known to be induced by interleukin-4 (IL4) and repressed by inflammatory cytokines, such as interleukin-1beta3 (IL1beta3) in human hepatocytes. The mechanisms involved in these transcriptional regulations remain elusive. In order to study these mechanisms, various constructs of the human CYP2E1 promoter were prepared and transfected into the human HepG2 hepatoma cell line. Our findings revealed that an IL4-responsive region of 128bp (-671/-544) was required to mediate induction by IL4. IL1beta caused moderate but significant decrease of the promoter activity, which was abolished when the two cytokines were combined. The IL1beta inhibitory effect is mediated through a regulatory sequence independent of that of IL4. Furthermore, by using specific signaling pathway inhibitors, we demonstrated that IL4 activation required protein kinase C (PKC) activation. In addition, our results suggest that induction by IL4 was not dependent on a single binding site but rather on a complex region which includes putative binding sites for signal transducer and activator of transcription (STAT)6, activator protein (AP)-1, nuclear factor kappa-B (NFkappaB), nuclear factor of activated T cells (NFAT) and CCAAT enhancer binding protein (C/EBP). Electrophoretic mobility shift assays suggest that AP1 and NFAT transcription factors are able to bind to three sites in the IL4-responsive region.
Collapse
|
925
|
Frazer-Abel AA, Baksh S, Fosmire SP, Willis D, Pierce AM, Meylemans H, Linthicum DS, Burakoff SJ, Coons T, Bellgrau D, Modiano JF. Nicotine activates nuclear factor of activated T cells c2 (NFATc2) and prevents cell cycle entry in T cells. J Pharmacol Exp Ther 2004; 311:758-69. [PMID: 15231866 DOI: 10.1124/jpet.104.070060] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We used primary peripheral blood T cells, a population that exists in G(0) and can be stimulated to enter the cell cycle synchronously, to define more precisely the effects of nicotine on pathways that control cell cycle entry and progression. Our data show that nicotine decreased the ability of T cells to transit through the G(0)/G(1) boundary (acquire competence) and respond to progression signals. These effects were due to nuclear factor of activated T cells c2 (NFATc2)-dependent repression of cyclin-dependent kinase 4 (CDK4) expression. Growth arrest at the G(0)/G(1) boundary was further enforced by inhibition of cyclin D2 expression and by increased expression and stabilization of p27Kip1. Intriguingly, T cells from habitual users of tobacco products and from NFATc2-deficient mice constitutively expressed CDK4 and were resistant to the antiproliferative effects of nicotine. These results indicate that nicotine impairs T cell cycle entry through NFATc2-dependent mechanisms and suggest that, in the face of chronic nicotine exposure, selection may favor cells that can evade these effects. We postulate that cross talk between nicotinic acetylcholine receptors and growth factor receptor-activated pathways offers a novel mechanism by which nicotine may directly impinge on cell cycle progression. This offers insight into possible reasons that underlie the unique effects of nicotine on distinct cell types and identifies new targets that may be useful control tobacco-related diseases.
Collapse
Affiliation(s)
- Ashley A Frazer-Abel
- Integrated Department of Immunology, University of Colorado Health Sciences Center, AMC Cancer Center, 2-Diamond Building, 1600 Pierce Street, Denver, CO 80214, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
926
|
Li H, Rao A, Hogan PG. Structural delineation of the calcineurin-NFAT interaction and its parallels to PP1 targeting interactions. J Mol Biol 2004; 342:1659-74. [PMID: 15364589 DOI: 10.1016/j.jmb.2004.07.068] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2004] [Revised: 07/16/2004] [Accepted: 07/20/2004] [Indexed: 01/21/2023]
Abstract
Calcineurin is a phosphoprotein phosphatase that channels intracellular Ca signals into multiple biological pathways. Calcineurin is known to interact directly with its substrate nuclear factor of activated T cells (NFAT or NFATc), with other substrates, and with several targeting and scaffold proteins including AKAP79 and Cabin1/cain. The calcineurin-NFAT interaction depends on recognition of a PxIxIT sequence motif present in NFAT-family proteins and in certain other calcineurin-interacting proteins. Here, we define the structural basis for the interaction of calcineurin with NFAT and with other proteins possessing the PxIxIT motif. The calcineurin-PxIxIT contact has a direct parallel in the contact of protein phosphatase 1 with its regulatory proteins, suggesting that the evolution of these related phosphatases involved local remodelling of an ancestral docking site.
Collapse
Affiliation(s)
- Huiming Li
- Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
927
|
Barlic J, McDermott DH, Merrell MN, Gonzales J, Via LE, Murphy PM. Interleukin (IL)-15 and IL-2 Reciprocally Regulate Expression of the Chemokine Receptor CX3CR1 through Selective NFAT1- and NFAT2-dependent Mechanisms. J Biol Chem 2004; 279:48520-34. [PMID: 15347678 DOI: 10.1074/jbc.m406978200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have recently reported that interleukin (IL)-15 and IL-2, which signal through IL-2Rbetagamma, oppositely regulate expression of the proinflammatory chemokine receptor CX3CR1. Here we delineate molecular mechanisms responsible for this paradox. By using a luciferase reporter plasmid, we identified a 433-bp region spanning the major transcriptional start point of human CX3CR1 that, when expressed in human peripheral blood mononuclear cells (PBMCs), possessed strong constitutive promoter activity. IL-2 and IL-15 treatment increased and abolished this activity, respectively, mimicking their effects on endogenous CX3CR1. IL-2 and IL-15 have been reported to also have opposite effects on the immunoregulatory transcription factor NFAT (nuclear factor of activated T cells), and the 433-bp region contains a kappaB-like NFAT site. The effects of IL-15 and IL-2 on both CX3CR1 reporter activity and endogenous CX3CR1 transcription in PBMCs were abolished by the NFAT inhibitors cyclosporin A and VIVIT. Moreover, mutation of the kappaB-like NFAT sequence markedly attenuated IL-2 and IL-15 modulation of CX3CR1 promoter-reporter activity in PBMCs. Furthermore, chromatin immunoprecipitation revealed that IL-15 promoted specific recruitment of NFAT1 but not NFAT2 to the CX3CR1 promoter, whereas IL-2 had the converse effect. This appears to be relevant in vivo because mouse CX3CR1 mRNA was expressed in both PBMCs and splenocytes from NFAT1-/- mice injected with recombinant IL-15 but was undetectable in cells from IL-15-injected NFAT1+/+ BALB/c mice; as predicted, IL-2 up-regulated cx3cr1 in both mouse strains to a similar extent. Thus, by pharmacologic, genetic, and biochemical criteria in vitro and in vivo, our results suggest that IL-15 and IL-2 oppositely regulate CX3CR1 gene expression by differentially recruiting NFAT1 and NFAT2 to a kappaB-like NFAT site within the CX3CR1 promoter. We propose that expression of CX3CR1 and possibly other immunoregulatory genes may be determined in part by the balance of NFAT1 and NFAT2 activity in leukocytes.
Collapse
MESH Headings
- Animals
- Base Sequence
- CX3C Chemokine Receptor 1
- Cell Line
- Chromatin/metabolism
- Chromatin Immunoprecipitation
- Crosses, Genetic
- DNA-Binding Proteins/metabolism
- Dose-Response Relationship, Drug
- Exons
- Gene Expression Regulation
- Genes, Reporter
- Humans
- Immunoprecipitation
- Interleukin-15/physiology
- Interleukin-2/metabolism
- Interleukin-2/physiology
- Leukocytes, Mononuclear/metabolism
- Luciferases/metabolism
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Models, Genetic
- Molecular Sequence Data
- Mutagenesis
- Mutation
- NFATC Transcription Factors
- Nuclear Proteins/metabolism
- Open Reading Frames
- Plasmids/metabolism
- Promoter Regions, Genetic
- Protein Structure, Tertiary
- RNA/metabolism
- Receptors, Chemokine/metabolism
- Recombinant Proteins/chemistry
- Time Factors
- Transcription Factors/metabolism
- Transcription, Genetic
- Transfection
- Up-Regulation
Collapse
Affiliation(s)
- Jana Barlic
- Molecular Signaling Section, Laboratory of Host Defenses, NIAID, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
928
|
Patel K, Muntoni F. Inducing muscle hypertrophy as a therapeutic strategy for muscular dystrophies. 122nd ENMC International Workshop, Naarden, The Netherlands, 28-30 November 2003. Neuromuscul Disord 2004; 14:519-25. [PMID: 15336693 DOI: 10.1016/j.nmd.2004.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Ketan Patel
- Veterinary Basic Sciences, Royal Veterinary College, Royal College Street, London, UK.
| | | |
Collapse
|
929
|
Johnson BV, Bert AG, Ryan GR, Condina A, Cockerill PN. Granulocyte-macrophage colony-stimulating factor enhancer activation requires cooperation between NFAT and AP-1 elements and is associated with extensive nucleosome reorganization. Mol Cell Biol 2004; 24:7914-30. [PMID: 15340054 PMCID: PMC515070 DOI: 10.1128/mcb.24.18.7914-7930.2004] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human granulocyte-macrophage colony-stimulating factor (GM-CSF) gene is activated by an NFAT-dependent enhancer forming an inducible DNase I hypersensitive (DH) site. The enhancer core comprising the DH site contains the GM330 and GM420 elements that bind NFAT and AP-1 cooperatively. Here we demonstrate that both elements are essential for enhancer activity and that Sp1 and AML1 sites in the enhancer become occupied in vivo only after activation. Chromatin structure analysis revealed that the GM-CSF enhancer core elements are divided between two adjacent nucleosomes that become destabilized and highly accessible after activation. Inducible chromatin reorganization was not restricted to the enhancer core but extended across a 3-kb domain of mobilized nucleosomes, within which the nucleosome repeat length was compressed from approximately 185 to 150 bp. The GM420 element is a high-affinity site that binds NFAT independently of AP-1 but depends on the linked AP-1 site for enhancer function. Nevertheless, just the NFAT motif from the GM420 element was sufficient to form a DH site within chromatin even in the absence of the AP-1 site. Hence, NFAT has the potential to cooperate with other transcription factors by promoting chromatin remodelling and increasing accessibility at inducible regulatory elements.
Collapse
Affiliation(s)
- Brett V Johnson
- Molecular Medicine Unit, Department of Medicine, St. James's University Hospital, University of Leeds, Leeds, England
| | | | | | | | | |
Collapse
|
930
|
Abstract
Experiments were initiated in avian embryos to determine the embryonic expression of calcineurin protein phosphatase isoforms as well as to identify developmental processes affected by inhibition of calcineurin signal transduction. Chicken calcineurin A alpha (CnAalpha) and calcineurin A beta (CnAbeta) are differentially expressed in the developing cardiovascular system, including primitive heart tube and valve primordia. Inhibition of calcineurin signaling by cyclosporin A (CsA) treatment in ovo resulted in distinct cardiovascular malformations, depending on the timing and localization of treatment. Initial formation of the heart tube was apparently normal in embryos treated with CsA from embryonic day (E)1 to E2, but hallmarks of heart failure were apparent with treatment from E2 to E3. Vascular defects were apparent in whole embryos treated on either day, but local administration of CsA directly to the forming vessels on E2 did not inhibit blood vessel formation. This observation supports an indirect effect of calcineurin inhibition on angiogenic remodeling as a result of compromised heart development. Together these studies are consistent with multiple roles for calcineurin signaling in the developing cardiovascular system.
Collapse
Affiliation(s)
- Christine M Liberatore
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, ML7020, Cincinnati, Ohio, USA
| | | |
Collapse
|
931
|
Xavier R, Rabizadeh S, Ishiguro K, Andre N, Ortiz JB, Wachtel H, Morris DG, Lopez-Ilasaca M, Shaw AC, Swat W, Seed B. Discs large (Dlg1) complexes in lymphocyte activation. ACTA ACUST UNITED AC 2004; 166:173-8. [PMID: 15263016 PMCID: PMC2172307 DOI: 10.1083/jcb.200309044] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
T cell antigen recognition involves the formation of a structured interface between antigen-presenting and T cells that facilitates the specific transmission of activating and desensitizing stimuli. The molecular machinery that organizes the signaling molecules and controls their disposition in response to activation remains poorly understood. We show here that in T cells Discs large (Dlg1), a PDZ domain-containing protein, is recruited upon activation to cortical actin and forms complexes with early participants in T cell activation. Transient overexpression of Dlg1 attenuates basal and Vav1-induced NFAT reporter activation. Reduction of Dlg1 expression by RNA interference enhances both CD3- and superantigen-mediated NFAT activation. Attenuation of antigen receptor signaling appears to be a complex, highly orchestrated event that involves the mutual segregation of important elements of the early signaling complex.
Collapse
Affiliation(s)
- Ramnik Xavier
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
932
|
Hoshino A, Matsumura S, Kondo K, Hirst JA, Fujii H. Inducible translocation trap: a system for detecting inducible nuclear translocation. Mol Cell 2004; 15:153-9. [PMID: 15225556 DOI: 10.1016/j.molcel.2004.06.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2004] [Revised: 05/03/2004] [Accepted: 05/14/2004] [Indexed: 11/30/2022]
Abstract
Here we report a general system, inducible translocation trap (ITT), for identification of proteins that translocate into the nucleus following signal transduction from cell surface receptors. ITT consists of a retroviral cDNA expression library of fusion proteins consisting of a LexA DNA binding domain, the transactivation domain of a transcriptional activator, and proteins encoded by cDNA inserts. The retroviral library is then transduced into cell lines containing a reporter gene with LexA binding sites in its promoter. Cells expressing the reporter gene by extracellular stimuli are then selected by flow-cytometric sorting. By using ITT, we identified cDNA encoding Stat1 in a screen of proteins which translocate into the nucleus by IFNgamma, indicating that this system can be used for isolation of nuclear translocating proteins induced by extracellular stimuli. ITT may be a useful tool for dissecting dynamic translocation in various biological systems.
Collapse
Affiliation(s)
- Akemi Hoshino
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
933
|
Ikeda F, Nishimura R, Matsubara T, Tanaka S, Inoue JI, Reddy SV, Hata K, Yamashita K, Hiraga T, Watanabe T, Kukita T, Yoshioka K, Rao A, Yoneda T. Critical roles of c-Jun signaling in regulation of NFAT family and RANKL-regulated osteoclast differentiation. J Clin Invest 2004; 114:475-84. [PMID: 15314684 PMCID: PMC503767 DOI: 10.1172/jci19657] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2003] [Accepted: 06/11/2004] [Indexed: 12/19/2022] Open
Abstract
Receptor activator of NF-kappaB ligand (RANKL) plays an essential role in osteoclast formation and bone resorption. Although genetic and biochemical studies indicate that RANKL regulates osteoclast differentiation by activating receptor activator of NF-kappaB and associated signaling molecules, the molecular mechanisms of RANKL-regulated osteoclast differentiation have not yet been fully established. We investigated the role of the transcription factor c-Jun, which is activated by RANKL, in osteoclastogenesis using transgenic mice expressing dominant-negative c-Jun specifically in the osteoclast lineage. We found that the transgenic mice manifested severe osteopetrosis due to impaired osteoclastogenesis. Blockade of c-Jun signaling also markedly inhibited soluble RANKL-induced osteoclast differentiation in vitro. Overexpression of nuclear factor of activated T cells 1 (NFAT1) (NFATc2/NFATp) or NFAT2 (NFATc1/NFATc) promoted differentiation of osteoclast precursor cells into tartrate-resistant acid phosphatase-positive (TRAP-positive) multinucleated osteoclast-like cells even in the absence of RANKL. Overexpression of NFAT1 also markedly transactivated the TRAP gene promoter. These osteoclastogenic activities of NFAT were abrogated by overexpression of dominant-negative c-Jun. Importantly, osteoclast differentiation and induction of NFAT2 expression by NFAT1 overexpression or soluble RANKL treatment were profoundly diminished in spleen cells of the transgenic mice. Collectively, these results indicate that c-Jun signaling in cooperation with NFAT is crucial for RANKL-regulated osteoclast differentiation.
Collapse
Affiliation(s)
- Fumiyo Ikeda
- Department of Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
934
|
Dixon LK, Abrams CC, Bowick G, Goatley LC, Kay-Jackson PC, Chapman D, Liverani E, Nix R, Silk R, Zhang F. African swine fever virus proteins involved in evading host defence systems. Vet Immunol Immunopathol 2004; 100:117-34. [PMID: 15207450 DOI: 10.1016/j.vetimm.2004.04.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
African swine fever virus (ASFV) can cause an acutely fatal haemorrhagic fever in domestic pigs although in its natural hosts, warthogs, bushpigs and the soft tick vector, Ornithodoros moubata, ASFV causes inapparent persistent infections. The virus is a large, cytoplasmic, double-stranded DNA virus which has a tropism for macrophages. As it is the only member of the Asfarviridae family, ASFV encodes many novel genes not encoded by other virus families. The ability of the virus to persist in its natural hosts and in domestic pigs, which recover from infection with less virulent isolates, shows that the virus has effective mechanisms to evade host defence systems. This review focuses on recent progress made in understanding the function of ASFV-encoded proteins, which are involved in modulating the host response to infection. Growing evidence suggests that a major strategy used by the virus is to modulate signalling pathways in infected macrophages, thus interfering with the expression of a large number of immunomodulatory genes. One potent immunomodulatory protein, A238L, inhibits both activation of the host NFkappaB transcription factor and inhibits calcineurin phosphatase activity. Calcineurin-dependent pathways, including activation of the NFAT transcription factor, are therefore inhibited. Another ASFV-encoded protein, CD2v, resembles the host CD2 protein, which is expressed on T cells and NK cells. This virus protein causes the adsorption of red blood cells around virus-infected cells and extracellular virus particles. Expression of the CD2v protein aids virus dissemination in pigs and the protein also has a role in impairing bystander lymphocyte function. This may be mediated either by a direct interaction of CD2v extracellular domain with ligands on lymphocytes or by an indirect mechanism involving interaction of the CD2v cytoplasmic tail with host proteins involved in signalling or trafficking pathways. Two ASFV proteins, an IAP and a Bcl2 homologue, inhibit apoptosis in infected cells and thus facilitate production of progeny virions. The prediction is that half to two-thirds of the approximately 150 genes encoded by ASFV are not essential for replication in cells but have an important role for virus survival and transmission in its hosts. These genes provide an untapped repository, and will be valuable tools for deciphering not only how the virus manipulates the host response to infection to avoid elimination, but also useful for understanding important host anti-viral mechanisms. In addition, they may provide leads for discovery of novel immunomodulatory drugs.
Collapse
Affiliation(s)
- Linda K Dixon
- Institute for Animal Health Pirbright Lab., Ash Road, Pirbright, Woking, Surrey GU24 ONF, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
935
|
Abstract
The apparent efficacy of B-cell depletion in autoimmune diseases has increased interest in targeting B cells. One goal of next generation therapies is to develop treatments that block B-cell activation and preserve resting nonautoimmune cells that maintain B cell memory. To do so, one needs to understand how B cells are activated and what receptors and intracellular signaling pathways regulate this process. This paper will summarize B-cell activation pathways and illustrate how these are being targeted in the development of new treatments for rheumatoid arthritis.
Collapse
Affiliation(s)
- Robert H Carter
- Department of Medicine, University of Alabama at Birmingham, 409 LHRB, 701 19th Street South, Birmingham, AL 35294, USA.
| |
Collapse
|
936
|
Im SH, Rao A. Activation and deactivation of gene expression by Ca2+/calcineurin-NFAT-mediated signaling. Mol Cells 2004; 18:1-9. [PMID: 15359117 DOI: 10.1016/s1016-8478(23)13074-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Ca2+/calcineurin-NFAT-mediated signaling pathways are involved in diverse cellular reactions by regulating gene expression either positively or negatively. The transcriptional activity of NFAT proteins can be either activating or deactivating depending on which binding partners are involved. Interaction of NFAT with AP-1 turns on the genes involved in active immune responses, while NFAT without cooperative binding of AP-1 turns on a T cell anergy program and blocks T cell activation and proliferation. In addition, interaction of NFAT with histone deacetylase (HDAC) proteins induces gene silencing. In this review we focus on the dual function, activator or deactivator, of NFAT and the binding partners that determine the role of NFAT in gene expression.
Collapse
Affiliation(s)
- Sin-Hyeog Im
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea.
| | | |
Collapse
|
937
|
Zaichuk TA, Shroff EH, Emmanuel R, Filleur S, Nelius T, Volpert OV. Nuclear factor of activated T cells balances angiogenesis activation and inhibition. ACTA ACUST UNITED AC 2004; 199:1513-22. [PMID: 15184502 PMCID: PMC2211785 DOI: 10.1084/jem.20040474] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
It has been demonstrated that vascular endothelial cell growth factor (VEGF) induction of angiogenesis requires activation of the nuclear factor of activated T cells (NFAT). We show that NFATc2 is also activated by basic fibroblast growth factor and blocked by the inhibitor of angiogenesis pigment epithelial–derived factor (PEDF). This suggests a pivotal role for this transcription factor as a convergence point between stimulatory and inhibitory signals in the regulation of angiogenesis. We identified c-Jun NH2-terminal kinases (JNKs) as essential upstream regulators of NFAT activity in angiogenesis. We distinguished JNK-2 as responsible for NFATc2 cytoplasmic retention by PEDF and JNK-1 and JNK-2 as mediators of PEDF-driven NFAT nuclear export. We identified a novel NFAT target, caspase-8 inhibitor cellular Fas-associated death domain–like interleukin 1β–converting enzyme inhibitory protein (c-FLIP), whose expression was coregulated by VEGF and PEDF. Chromatin immunoprecipitation showed VEGF-dependent increase of NFATc2 binding to the c-FLIP promoter in vivo, which was attenuated by PEDF. We propose that one possible mechanism of concerted angiogenesis regulation by activators and inhibitors may be modulation of the endothelial cell apoptosis via c-FLIP controlled by NFAT and its upstream regulator JNK.
Collapse
Affiliation(s)
- Tetiana A Zaichuk
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | |
Collapse
|
938
|
Ikeda F, Nishimura R, Matsubara T, Tanaka S, Inoue JI, Reddy SV, Hata K, Yamashita K, Hiraga T, Watanabe T, Kukita T, Yoshioka K, Rao A, Yoneda T. Critical roles of c-Jun signaling in regulation of NFAT family and RANKL-regulated osteoclast differentiation. J Clin Invest 2004. [DOI: 10.1172/jci200419657] [Citation(s) in RCA: 350] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
939
|
Matsumoto M, Kogawa M, Wada S, Takayanagi H, Tsujimoto M, Katayama S, Hisatake K, Nogi Y. Essential role of p38 mitogen-activated protein kinase in cathepsin K gene expression during osteoclastogenesis through association of NFATc1 and PU.1. J Biol Chem 2004; 279:45969-79. [PMID: 15304486 DOI: 10.1074/jbc.m408795200] [Citation(s) in RCA: 347] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The receptor activator of NF-kappaB ligand (RANKL) induces various osteoclast-specific marker genes during osteoclast differentiation mediated by mitogen-activated protein (MAP) kinase cascades. However, the results of transcriptional programming of an osteoclast-specific cathepsin K gene are inconclusive. Here we report the regulatory mechanisms of RANKL-induced cathepsin K gene expression during osteoclastogenesis in a p38 MAP kinase-dependent manner. The reporter gene analysis with sequential 5'-deletion constructs of the cathepsin K gene promoter indicates that limited sets of the transcription factors such as NFATc1, PU.1, and microphthalmia transcription factor indeed enhance synergistically the gene expression when overexpressed in RAW264 cells. In addition, the activation of p38 MAP kinase is required for the maximum enhancement of the gene expression. RANKL-induced NFATc1 forms a complex with PU.1 in nuclei of osteoclasts following the nuclear accumulation of NFATc1 phosphorylated by the activated p38 MAP kinase. These results suggest that the RANKL-induced cathepsin K gene expression is cooperatively regulated by the combination of the transcription factors and p38 MAP kinase in a gradual manner.
Collapse
Affiliation(s)
- Masahito Matsumoto
- Department of Molecular Biology, Saitama Medical School, Saitama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
940
|
Ahamed J, Venkatesha RT, Thangam EB, Ali H. C3a enhances nerve growth factor-induced NFAT activation and chemokine production in a human mast cell line, HMC-1. THE JOURNAL OF IMMUNOLOGY 2004; 172:6961-8. [PMID: 15153516 DOI: 10.4049/jimmunol.172.11.6961] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of cell surface G protein-coupled receptors leads to transphosphorylation and activation of a number of receptor tyrosine kinases. Human mast cells express G protein-coupled receptors for the complement component C3a (C3aR) and high affinity nerve growth factor (NGF) receptor tyrosine kinase, TrkA. To determine whether C3a cross-regulates TrkA signaling and biological responses, we used a human mast cell-line, HMC-1, that natively expresses both receptors. We found that NGF caused tyrosine phosphorylation of TrkA, resulting in a sustained Ca(2+) mobilization, NFAT activation, extracellular-signal regulated kinase (ERK) phosphorylation, and chemokine, macrophage inflammatory protein-1beta (MIP-1beta) production. In contrast, C3a induced a transient Ca(2+) mobilization and ERK phosphorylation but failed to stimulate TrkA phosphorylation, NFAT activation, or MIP-1beta production. Surprisingly, C3a significantly enhanced NGF-induced NFAT activation, ERK phosphorylation, and MIP-1beta production. Pertussis toxin, a G(i/o) inhibitor, selectively blocked priming by C3a but had no effect on NGF-induced responses. Mitogen-activated protein/ERK kinase inhibitor U0126 caused approximately 30% inhibition of NGF-induced MIP-1beta production but had no effect on priming by C3a. However, cyclosporin A, an inhibitor of calcineurin-mediated NFAT activation, caused substantial inhibition of NGF-induced MIP-1beta production both in the absence and presence of C3a. These data demonstrate that NGF caused tyrosine phosphorylation of TrkA to induce chemokine production in HMC-1 cells via a pathway that mainly depends on sustained Ca(2+) mobilization and NFAT activation. Furthermore, C3a enhances NGF-induced transcription factor activation and chemokine production via a G protein-mediated pathway that does not involve TrkA phosphorylation.
Collapse
Affiliation(s)
- Jasimuddin Ahamed
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
941
|
Kiani A, Habermann I, Haase M, Feldmann S, Boxberger S, Sanchez-Fernandez MA, Thiede C, Bornhäuser M, Ehninger G. Expression and regulation of NFAT (nuclear factors of activated T cells) in human CD34+cells: down-regulation upon myeloid differentiation. J Leukoc Biol 2004; 76:1057-65. [PMID: 15292278 DOI: 10.1189/jlb.0404259] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The calcineurin-dependent, cyclosporin A (CsA)-sensitive transcription factor nuclear factor of activated T cells (NFAT) represents a group of proteins, which is well-characterized as a central regulatory element of cytokine expression in activated T cells. In contrast, little is known about the expression or function of NFAT family members in myeloid cells; moreover, it is unclear whether they are expressed by hematopoietic stem/progenitor cells. Here, we show that NFATc2 (NFAT1) is expressed at high levels in CD34+ cells and megakaryocytes but not in cells committed to the neutrophilic, monocytic, or erythroid lineages. Cytokine-induced in vitro differentiation of CD34+ cells into neutrophil granulocytes results in the rapid suppression of NFATc2 RNA and protein. NFATc2 dephosphorylation/rephosphorylation as well as nuclear/cytoplasmic translocation in CD34+ cells follow the same calcineurin-dependent pattern as in T lymphocytes, suggesting that NFATc2 activation in these cells is equally sensitive to inhibition with CsA. Finally, in vitro proliferation, but not differentiation, of CD34+ cells cultured in the presence of fms-like tyrosine kinase 3 ligand (FLT3L), stem cell factor, granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin-3, and G-CSF is profoundly inhibited by treatment with CsA in a dose-dependent manner. These results suggest a novel and unexpected role for members of the NFAT transcription factor family in the hematopoietic system.
Collapse
Affiliation(s)
- Alexander Kiani
- Department of Medicine I, University Hospital Carl Gustav Carus, University of Dresden Technical Center, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
942
|
Davé V, Childs T, Whitsett JA. Nuclear Factor of Activated T Cells Regulates Transcription of the Surfactant Protein D Gene (Sftpd) via Direct Interaction with Thyroid Transcription Factor-1 in Lung Epithelial Cells. J Biol Chem 2004; 279:34578-88. [PMID: 15173172 DOI: 10.1074/jbc.m404296200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Surfactant protein D (SP-D) plays critical roles in host defense, surfactant homeostasis, and pulmonary immunomodulation. Here, we identify a role of nuclear factor of activated T cells (NFATs) in regulation of murine SP-D gene (Sftpd) transcription. An NFAT-dependent enhancer modulated by NFATs or calcineurin and sensitive to cyclosporin was identified in the Sftpd promoter. Ionomycin and phorbol 12-myristate 13-acetate further increased the activity of this enhancer, whereas VIVIT, a potent NFAT inhibitor peptide, selectively interfered with the calcineurin-NFAT interaction and abolished enhancer function. Gel supershift and DNase I protection assays identified DNA elements that bind NFAT in the Sftpd promoter. Calcineurin and NFATc3 proteins were detected in the embryonic and adult mouse lung epithelium, and the mRNA expression profiles of the NFATs were similar in immortalized mouse lung epithelial cells and alveolar epithelial type II cells. NFATc3 and TTF-1 activated the Sftpd promoter, synergized transcription, co-immunoprecipitated from mouse lung epithelial cells, and physically interacted in vitro. Components of the calcineurin/NFAT pathway were identified in respiratory epithelial cells of the lung that potentially augment rapid assembly of a multiprotein transcription complex on Sftpd promoter inducing SP-D expression.
Collapse
Affiliation(s)
- Vrushank Davé
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| | | | | |
Collapse
|
943
|
McCullagh KJA, Calabria E, Pallafacchina G, Ciciliot S, Serrano AL, Argentini C, Kalhovde JM, Lømo T, Schiaffino S. NFAT is a nerve activity sensor in skeletal muscle and controls activity-dependent myosin switching. Proc Natl Acad Sci U S A 2004; 101:10590-5. [PMID: 15247427 PMCID: PMC489979 DOI: 10.1073/pnas.0308035101] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2003] [Indexed: 12/27/2022] Open
Abstract
Calcineurin (Cn) signaling has been implicated in nerve activity-dependent fiber type specification in skeletal muscle, but the downstream effector pathway has not been established. We have investigated the role of the transcription factor nuclear factor of activated T cells (NFAT), a major target of Cn, by using an in vivo transfection approach in regenerating and adult rat muscles. NFAT transcriptional activity was monitored with two different NFAT-dependent reporters and was found to be higher in slow compared to fast muscles. NFAT activity is decreased by denervation in slow muscles and is increased by electrostimulation of denervated muscles with a tonic low-frequency impulse pattern, mimicking the firing pattern of slow motor neurons, but not with a phasic high-frequency pattern typical of fast motor neurons. To determine the role of NFAT, we transfected regenerating and adult rat muscles with a plasmid coding for VIVIT, a specific peptide inhibitor of Cn-mediated NFAT activation. VIVIT was found to block the expression of slow myosin heavy chain (MyHC-slow) induced by slow motor neuron activity in regenerating slow soleus muscle and to inhibit the expression of MyHC-slow transcripts and the activity of a MyHC-slow promoter in adult soleus. The role of NFAT was confirmed by the finding that a constitutively active NFATc1 mutant stimulates the MyHC-slow, inhibits the fast MyHC-2B promoter in adult fast muscles, and induces MyHC-slow expression in regenerating muscles. These results support the notion that Cn-NFAT signaling acts as a nerve activity sensor in skeletal muscle in vivo and controls nerve activity-dependent myosin switching.
Collapse
Affiliation(s)
- Karl J A McCullagh
- Department of Biomedical Sciences, Consiglio Nazionale delle Ricerche Institute of Neurosciences, University of Padua, 35121 Padua, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
944
|
Hesser BA, Liang XH, Camenisch G, Yang S, Lewin DA, Scheller R, Ferrara N, Gerber HP. Down syndrome critical region protein 1 (DSCR1), a novel VEGF target gene that regulates expression of inflammatory markers on activated endothelial cells. Blood 2004; 104:149-58. [PMID: 15016650 DOI: 10.1182/blood-2004-01-0273] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Abstract
We conducted a genome-wide analysis of genes that are regulated by vascular endothelial growth factor (VEGF) in endothelial cells and identified DSCR1 to be most significantly induced. Consistent with an antagonistic function on calcineurin (CnA) signaling, expression of DSCR1 in endothelial cells blocked dephosphorylation, nuclear translocation, and activity of nuclear factor of activated T cell (NFAT), a transcription factor involved in mediating CnA signaling. DSCR1 was not only induced by VEGF, but also by other compounds activating CnA signaling, suggesting a more general role for DSCR1 in activated endothelial cells. Transient expression of DSCR1 attenuated inflammatory marker genes such as tissue factor (TF), E-selectin, and Cox-2, identifying a previously unknown regulatory role for DSCR1 in activated endothelial cells. In contrast, knock-down of endogenous DSCR1 increased NFAT activity and stimulated expression of inflammatory genes on activated endothelial cells. Thus, the negative regulatory feedback loop between DSCR1 and CnA signaling in endothelial cells identified may represent a potential molecular mechanism underlying the frequently transient expression of inflammatory genes following activation of endothelial cells.
Collapse
Affiliation(s)
- Boris A Hesser
- Department of Molecular Oncology, Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | |
Collapse
|
945
|
Serfling E, Berberich-Siebelt F, Avots A, Chuvpilo S, Klein-Hessling S, Jha MK, Kondo E, Pagel P, Schulze-Luehrmann J, Palmetshofer A. NFAT and NF-κB factors—the distant relatives. Int J Biochem Cell Biol 2004; 36:1166-70. [PMID: 15109564 DOI: 10.1016/j.biocel.2003.07.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2003] [Revised: 07/28/2003] [Accepted: 07/31/2003] [Indexed: 11/24/2022]
Abstract
NFAT and NF-kappaB proteins are members of a superfamily of transcription factors whose activity plays a crucial role in the activation, proliferation and apoptosis of lymphocytes. Both types of factors share a number of properties, including similar DNA binding domains and rapid nuclear translocation upon antigenic stimulation. While NF-kappaBs control both innate and adaptive immune responses, NFATs control the adaptive immune system which emerged-in parallel with the appearance of the NFAT family-in jawed fish. However, NFATs and NF-kappaBs differ remarkably in their function. Whereas NFATs support activation-induced cell death (AICD) of T and B cells, NF-kappaB proteins frequently exert a strong anti-apoptotic effect on lymphocytes and other cells. While the anti-apoptotic activity of NF-kappaBs contributes to their oncogenic capacity, the pro-apoptotic activity favors NFATs as tumor suppressors in lymphoid cells.
Collapse
Affiliation(s)
- Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Josef-Schneider-Str. 2, D-97080 Würzburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
946
|
Monticelli S, Solymar DC, Rao A. Role of NFAT proteins in IL13 gene transcription in mast cells. J Biol Chem 2004; 279:36210-8. [PMID: 15229217 DOI: 10.1074/jbc.m406354200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Th2 and mast cells are participants in the asthmatic response to allergens, and both cell types produce the cytokines interleukin (IL)-4 and IL-13. IL-13 in particular is both necessary and sufficient for experimental models of asthma. The transcription factor NFAT plays a central role in cytokine transcriptional regulation in both cell types. Here, we analyze the molecular basis of IL13 gene transcription in Th2 and mast cells. We show that NFAT1 is the major NFAT protein involved in regulating IL13 transcription in mast cells. Although NFAT2 is correctly expressed and regulated in mast cells, it does not contribute to IL13 gene transcription as shown by analysis of cells lacking NFAT2 and cells expressing a constitutively active version of NFAT2. The difference between NFAT1 and NFAT2 appears to be due to a preferential synergistic interaction of NFAT1 with GATA proteins at the IL13 promoter. We suggest that mast cells lack a co-activator protein that stabilizes the binding of NFAT2 to the IL13 promoter by interacting either with NFAT2 itself or with a DNA-bound complex of NFAT2 and GATA proteins.
Collapse
Affiliation(s)
- Silvia Monticelli
- Department of Pathology, Harvard Medical School, and CBR Institute for Biomedical Research, Boston, MA 02115, USA
| | | | | |
Collapse
|
947
|
Okamura H, Garcia-Rodriguez C, Martinson H, Qin J, Virshup DM, Rao A. A conserved docking motif for CK1 binding controls the nuclear localization of NFAT1. Mol Cell Biol 2004; 24:4184-95. [PMID: 15121840 PMCID: PMC400483 DOI: 10.1128/mcb.24.10.4184-4195.2004] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In resting cells, the NFAT1 transcription factor is kept inactive in the cytoplasm by phosphorylation on multiple serine residues. These phosphorylated residues are primarily contained within two types of serine-rich motifs, the SRR-1 and SP motifs, which are conserved within the NFAT family. Several different kinases have been proposed to regulate NFAT, but no single candidate displays the specificity required to fully phosphorylate both types of motifs; thus, the identity of the kinase that regulates NFAT activity remains unclear. Here we show that the NFAT1 serine motifs are regulated by distinct kinases that must coordinate to control NFAT1 activation. CK1 phosphorylates only the SRR-1 motif, the primary region required for NFAT1 nuclear import. CK1 exists with NFAT1 in a high-molecular-weight complex in resting T cells but dissociates upon activation. GSK3 does not phosphorylate the SRR-1 region but can target the NFAT1 SP-2 motif, and it synergizes with CK1 to regulate NFAT1 nuclear export. We identify a conserved docking site for CK1 in NFAT proteins and show that mutation of this site disrupts NFAT1-CK1 interaction and causes constitutive nuclear localization of NFAT1. The CK1 docking motif is present in proteins of the Wnt, Hedgehog, and circadian-rhythm pathways, which also integrate the activities of CK1 and GSK3.
Collapse
Affiliation(s)
- Heidi Okamura
- CBR Institute for Biomedical Research and Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
948
|
Choi JY, Beaman-Hall CM, Vallano ML. Granule neurons in cerebellum express distinct splice variants of the inositol trisphosphate receptor that are modulated by calcium. Am J Physiol Cell Physiol 2004; 287:C971-80. [PMID: 15189817 DOI: 10.1152/ajpcell.00571.2003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Primary cultures of granule cells (GC) from rat cerebellar cortex were used to determine whether bioelectric activity, via a Ca(2+)/calmodulin-dependent kinase (CaMK) signaling cascade, modulates expression and exon selection in the inositol trisphosphate receptor type 1 (IP(3)R1). IP(3)R1 contains or lacks three exons (S1, S2, and S3) that are regulated in a regionally and temporally specific manner. The neuronal, or long, form of IP(3)R1 is distinguished from peripheral tissues by inclusion of the S2 exon. Although previous studies indicated that IP(3)R1 are undetectable in the cerebellar granular layer in vivo, receptor protein and mRNA are induced in cultured GC grown in medium supplemented with 25 mM KCl or NMDA, two trophic agents that promote long-term survival, compared with GC grown in 5 mM KCl. IP(3)R1 induction in response to 25 mM KCl or NMDA is attenuated by coaddition of voltage-sensitive calcium channel or NMDA receptor antagonists, respectively. Actinomycin D, CaMK, and calcineurin antagonists likewise suppress induction. Unlike the major variants of IP(3)R1 in Purkinje neurons, which lack S1 and S3, GC grown with trophic agents express mRNA containing these exons. Both neuronal types contain S2. Evidence obtained using mutant mice with Purkinje cell lesions, laser-microdissected GC neurons from slices, and explant cultures indicates that GC predominantly express the S1-containing variant of IP(3)R1 in vivo.
Collapse
Affiliation(s)
- Joseph Y Choi
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | | | | |
Collapse
|
949
|
Venkatesh N, Feng Y, DeDecker B, Yacono P, Golan D, Mitchison T, McKeon F. Chemical genetics to identify NFAT inhibitors: potential of targeting calcium mobilization in immunosuppression. Proc Natl Acad Sci U S A 2004; 101:8969-74. [PMID: 15184684 PMCID: PMC428456 DOI: 10.1073/pnas.0402803101] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The development of more selective immunosuppressive agents to mitigate transplant rejection and autoimmune diseases requires effective strategies of blocking signaling pathways in T cells. Current immunosuppressive strategies use cyclosporin A (CsA) or FK506 to inhibit calcineurin, which dephosphorylates and promotes the nuclear import of nuclear factor of activated T cells (NFAT) transcription factors. These nuclear NFATs then transactivate cytokine genes that regulate proliferative responses of T cells. Both CsA and FK506 have debilitating side effects, including nephrotoxicity, hypertension, diabetes, and seizures, that argue for the development of alternative or complementary agents. To this end, we developed cell-based assays for monitoring NFAT dynamics in nonlymphoid cells to identify small molecules that inhibit NFAT nuclear import. Interestingly, we found that the majority of these small molecules suppress NFAT signaling by interfering with "capacitative" or "store-operated" calcium mobilization, thus raising the possibility that such mobilization processes are relevant targets in immunosuppression therapy. Further, these small molecules also show dose-dependent suppression of cytokine gene expression in T cells. Significantly, the IC(50) of CsA in primary T cells was reduced by the addition of suboptimal concentrations of these compounds, suggesting the possibility that such small molecules, in combination with CsA, offer safer means of immunosuppression.
Collapse
|
950
|
Jordan BA, Fernholz BD, Boussac M, Xu C, Grigorean G, Ziff EB, Neubert TA. Identification and verification of novel rodent postsynaptic density proteins. Mol Cell Proteomics 2004; 3:857-71. [PMID: 15169875 DOI: 10.1074/mcp.m400045-mcp200] [Citation(s) in RCA: 243] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The postsynaptic density (PSD) is a cellular structure specialized in receiving and transducing synaptic information. Here we describe the identification of 452 proteins isolated from biochemically purified PSD fractions of rat and mouse brains using nanoflow HPLC coupled to electrospray tandem mass spectrometry (LC-MS/MS). Fluorescence microscopy and Western blotting were used to verify that many of the novel proteins identified exhibit subcellular distributions consistent with those of PSD-localized proteins. In addition to identifying most previously described PSD components, we also detected proteins involved in signaling to the nucleus as well as regulators of ADP-ribosylation factor signaling, ubiquitination, RNA trafficking, and protein translation. These results suggest new mechanisms by which the PSD helps regulate synaptic strength and transmission.
Collapse
Affiliation(s)
- Bryen A Jordan
- Department of Biochemistry, New York University, School of Medicine, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|