1
|
Tapia C, Principe G, González-Pardo V. 1α,25(OH) 2D 3 regulates pro-angiogenic factors in endothelial cells transformed by Kaposi's sarcoma-associated herpesvirus G protein coupled receptor. Biochimie 2023; 212:76-84. [PMID: 37062469 DOI: 10.1016/j.biochi.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/01/2023] [Accepted: 04/05/2023] [Indexed: 04/18/2023]
Abstract
When tumoral cell expansion exceeds the vascular supply, regions of hypoxia or low oxygen concentration are generated promoting the formation of new vessels through cell proliferation and migration. Viral G protein-coupled receptor (vGPCR) is associated to Kaposi's sarcoma pathology and induces a paracrine transformation when is stably expressed in murine endothelial cells activating hypoxia-induced transcription factors. Previously, we reported the antiproliferative actions of 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) in endothelial cells transformed by the vGPCR (SVEC-vGPCR). Herein, we further investigated if pro-angiogenic factors as AP-1, HIF-1α and VEGF are modulated by 1α,25(OH)2D3. We found by qRT-PCR analysis that the mRNA level of JunB, a negative regulator of cell proliferation, was similarly increased at all-time points tested after 1α,25(OH)2D3 treatment in SVEC-vGPCR cells. Also, mRNA levels of the pro-angiogenic factor c-Fos, which induces tumor invasion, were only decreased during one short period treatment. In addition, Hif-1α mRNA and protein levels were significantly reduced after 1α,25(OH)2D3 treatment in a VDR dependent fashion. However, mRNA levels of the angiogenic activator Vegf, promoted in turn by Hif-1α expression, were surprisingly high depending on VDR expression as well. Moreover, Egr-1, which has been reported to induce VEGF expression independently of HIF-1α, diminished its expression with 1α,25(OH)2D3 treatment, fact that was related to the decline of p-ERK1/2. Altogether, these results suggest a negative modulation of some pro-angiogenic factors like AP-1 and HIF-1α, as part of the antiproliferative mechanism of 1α,25(OH)2D3 in SVEC-vGPCR endothelial cells.
Collapse
Affiliation(s)
- Cinthya Tapia
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia-Universidad Nacional del Sur (UNS), Argentina
| | - Gabriel Principe
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia-Universidad Nacional del Sur (UNS), Argentina
| | - Verónica González-Pardo
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia-Universidad Nacional del Sur (UNS), Argentina.
| |
Collapse
|
2
|
Zheng X, Sun L, Liu B, Huang Z, Zhu Y, Chen T, Jia L, Li Y, Lei W. Morphological Study of the Cortical and Thalamic Glutamatergic Synaptic Inputs of Striatal Parvalbumin Interneurons in Rats. Neurochem Res 2021; 46:1659-1673. [PMID: 33770320 DOI: 10.1007/s11064-021-03302-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 02/14/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Parvalbumin-immunoreactive (Parv+) interneurons is an important component of striatal GABAergic microcircuits, which receive excitatory inputs from the cortex and thalamus, and then target striatal projection neurons. The present study aimed to examine ultrastructural synaptic connection features of Parv+ neruons with cortical and thalamic input, and striatal projection neurons by using immuno-electron microscopy (immuno-EM) and immunofluorescence techniques. Our results showed that both Parv+ somas and dendrites received numerous asymmetric synaptic inputs, and Parv+ terminals formed symmetric synapses with Parv- somas, dendrites and spine bases. Most interestingly, spine bases targeted by Parv+ terminals simultaneously received excitatory inputs at their heads. Electrical stimulation of the motor cortex (M1) induced higher proportion of striatal Parv+ neurons express c-Jun than stimulation of the parafascicular nucleus (PFN), and indicated that cortical- and thalamic-inputs differentially modulate Parv+ neurons. Consistent with that, both Parv + soma and dendrites received more VGlut1+ than VGlut2+ terminals. However, the proportion of VGlut1+ terminal targeting onto Parv+ proximal and distal dendrites was not different, but VGlut2+ terminals tended to target Parv+ somas and proximal dendrites than distal dendrites. These functional and morphological results suggested excitatory cortical and thalamic glutamatergic inputs differently modulate Parv+ interneurons, which provided inhibition inputs onto striatal projection neurons. To maintain the balance between the cortex and thalamus onto Parv+ interneurons may be an important therapeutic target for neurological disorders.
Collapse
Affiliation(s)
- Xuefeng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou, China
| | - Liping Sun
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bingbing Liu
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yaofeng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Institute of Medicine, College of Medicine, Jishou University, Jishou, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Linju Jia
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yanmei Li
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
3
|
Wang H, Shi C, Kong M, Mu C, Wei H, Wang C. Cloning and expression of a transcription factor activator protein-1 member identified from the swimming crab Portunus trituberculatus. Cell Stress Chaperones 2018; 23:1275-1282. [PMID: 30255490 PMCID: PMC6237694 DOI: 10.1007/s12192-018-0935-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/15/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022] Open
Abstract
Transcription activator proteins are regulatory proteins that bind to the promoter regions of target genes. Transcription activator protein-1 (AP-1) regulates numerous genes related to the immune system, apoptosis, and proliferation. In this study, the full-length cDNA of AP-1 from Portunus trituberculatus (PtAP-1) was identified by expressed sequence tag analysis and cDNA-end rapid amplification. The gene is 1183 bp and encodes a 256-amino acid protein with a predicted molecular mass and isoelectric point of 28.96 kDa and 8.90, respectively. PtAP-1 showed the highest expression level in the gonad tissue and the lowest expression level in blood, hemocyte, muscle, hepatopancreas, and gill, during the first 6 h of low-salinity stimulation (10%). Additionally, we observed steady decreases in PtAP-1 mRNA expression in the gill, but at 12 h, expression was initially upregulated, followed by a significant decrease until restoration to baseline levels at 48 h. Additionally, Vibrio alginolyticus challenge resulted in significant upregulation of PtAP-1 expression in the first 6 h, which was maintained at high levels for 48 h. From 48 to 72 h, we observed decreases in PtAP-1 levels, although they remained significantly higher than those detected at baseline. These results suggested that PtAP-1 is involved in the immune response and osmoregulation of crustaceans.
Collapse
Affiliation(s)
- Huan Wang
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo, 315211 People’s Republic of China
| | - Ce Shi
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo, 315211 People’s Republic of China
| | - Mengyao Kong
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
| | - Changkao Mu
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo, 315211 People’s Republic of China
| | - Hongling Wei
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
| | - Chunlin Wang
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Faculty of Life Science and Biotechnology, Ningbo University, Ningbo, 315211 People’s Republic of China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo, 315211 People’s Republic of China
| |
Collapse
|
4
|
Gao XQ, Ge YS, Shu QH, Ma HX. Expression of Fra-1 in human hepatocellular carcinoma and its prognostic significance. Tumour Biol 2017; 39:1010428317709635. [PMID: 28653890 DOI: 10.1177/1010428317709635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This study aimed to explore the clinical significance and prognostic value of Fra-1 in hepatocellular carcinoma patients after curative resection. Fra-1 expression was investigated using a combination of techniques: immunohistochemistry for 66 samples of hepatocellular carcinoma and quantitative real-time polymerase chain reaction and western blotting assays for 19 matched hepatocellular carcinoma specimens. Fra-1 was present in 38 of 66 (57.6%) tumor tissues, with intense staining in the nuclei. There was also positive staining in 14 of 66 (21.2%) adjacent peritumoral tissues, with weak staining in the cytoplasm. Quantitative real-time polymerase chain reaction and western blotting assays confirmed higher expression of Fra-1 messenger RNA and Fra-1 protein in tumor tissues than adjacent non-tumor tissues for 19 hepatocellular carcinoma samples (p < 0.001). Positive expression of Fra-1 was significantly related to vascular invasion and serum alpha-fetoprotein. Kaplan-Meier survival analysis found that overexpressed Fra-1 was correlated with poor overall survival and disease-free survival. Multivariate analysis identified Fra-1 as an independent prognostic factor. Fra-1 may be involved in the progress of hepatocellular carcinoma and could be a promising molecular candidate in the diagnosis and treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiao-Qiang Gao
- 1 Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery and Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Yong-Sheng Ge
- 1 Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery and Department of Hepatic Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Qing-Hua Shu
- 2 Department of General Surgery, Nanjing Second People's Hospital, Nanjing, China
| | - Hua-Xing Ma
- 3 Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| |
Collapse
|
5
|
Rawat V, Goux W, Piechaczyk M, D Mello SR. c-Fos Protects Neurons Through a Noncanonical Mechanism Involving HDAC3 Interaction: Identification of a 21-Amino Acid Fragment with Neuroprotective Activity. Mol Neurobiol 2015; 53:1165-1180. [PMID: 25592718 DOI: 10.1007/s12035-014-9058-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/08/2014] [Indexed: 11/26/2022]
Abstract
Proteins belonging to the AP-1 family of transcription factors are known to be involved in the regulation of neuronal viability. While strides have been made to elucidate the mechanisms of how individual members regulate cell death, much remains unknown. We find that the expression of one AP-1 member, c-Fos, is reduced in cerebellar granule neurons (CGNs) induced to die by low potassium (LK) treatment. Restoration and increase of this expression protect CGNs against LK-induced death, whereas knockdown induces death of otherwise healthy neurons. Furthermore, forced expression can protect cortical neurons against homocysteic acid (HCA)-induced toxicity. Taken together, this suggests that c-Fos is necessary for neuronal survival and that elevating c-Fos expression has a neuroprotective effect. Consistent with this idea is the finding that c-Fos expression is reduced selectively in the striatum in two separate mouse models of Huntington's disease and forced expression protects against neuronal death resulting from mutant huntingtin (mut-Htt) expression. Interestingly, neuroprotection by c-Fos does not require its DNA-binding, transcriptional, or heteromerization domains. However, this protective activity can be inhibited by pharmacological inhibition of c-Abl, CK-I, and MEK-ERK signaling. Additionally, expression of point mutant forms of this protein has identified that mutation of a tyrosine residue, Tyr345, can convert c-Fos from neuroprotective to neurotoxic. We show that c-Fos interacts with histone deacetylase-3 (HDAC3), a protein that contributes to mut-Htt neurotoxicity and whose overexpression is sufficient to promote neuronal death. When co-expressed, c-Fos can protect against HDAC3 neurotoxicity. Finally, our study identifies a 21-amino acid region at the C-terminus of c-Fos that is sufficient to protect neurons against death induced by LK, HCA treatment, or mut-Htt expression when expressed via a plasmid transfection or as a cell-permeable peptide. This cell-permeable peptide, designated as Fos-CTF, could have potential as a therapeutic agent for neurodegenerative diseases.
Collapse
Affiliation(s)
- Varun Rawat
- Department of Biological Sciences, Southern Methodist University, Dedman Life Sciences Building, 6501 Airline Road, Dallas, TX, 75275, USA
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Warren Goux
- Department of Chemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Santosh R D Mello
- Department of Biological Sciences, Southern Methodist University, Dedman Life Sciences Building, 6501 Airline Road, Dallas, TX, 75275, USA.
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
6
|
Healy S, Khan P, Davie JR. Immediate early response genes and cell transformation. Pharmacol Ther 2013; 137:64-77. [DOI: 10.1016/j.pharmthera.2012.09.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 09/06/2012] [Indexed: 01/20/2023]
|
7
|
Tan LJ, Saijo M, Kuraoka I, Narita T, Takahata C, Iwai S, Tanaka K. Xeroderma pigmentosum group F protein binds to Eg5 and is required for proper mitosis: implications for XP-F and XFE. Genes Cells 2012; 17:173-85. [PMID: 22353549 DOI: 10.1111/j.1365-2443.2012.01582.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The xeroderma pigmentosum group F-cross-complementing rodent repair deficiency group 1 (XPF-ERCC1) complex is a structure-specific endonuclease involved in nucleotide excision repair (NER) and interstrand cross-link (ICL) repair. Patients with XPF mutations may suffer from two forms of xeroderma pigmentosum (XP): XP-F patients show mild photosensitivity and proneness to skin cancer but rarely show any neurological abnormalities, whereas XFE patients display symptoms of severe XP symptoms, growth retardation and accelerated aging. Xpf knockout mice display accelerated aging and die before weaning. These results suggest that the XPF-ERCC1 complex has additional functions besides NER and ICL repair and is essential for development and growth. In this study, we show a partial colocalization of XPF with mitotic spindles and Eg5. XPF knockdown in cells led to an increase in the frequency of abnormal nuclear morphology and mitosis. Similarly, the frequency of abnormal nuclei and mitosis was increased in XP-F and XFE cells. In addition, we showed that Eg5 enhances the action of XPF-ERCC1 nuclease activity. Taken together, these results suggest that the interaction between XPF and Eg5 plays a role in mitosis and DNA repair and offer new insights into the pathogenesis of XP-F and XFE.
Collapse
Affiliation(s)
- Li Jing Tan
- Human Cell Biology Group, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
8
|
Effects of site-directed mutagenesis of mglA on motility and swarming of Myxococcus xanthus. BMC Microbiol 2010; 10:295. [PMID: 21083931 PMCID: PMC3000849 DOI: 10.1186/1471-2180-10-295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 11/18/2010] [Indexed: 01/31/2023] Open
Abstract
Background The mglA gene from the bacterium Myxococcus xanthus encodes a 22kDa protein related to the Ras superfamily of monomeric GTPases. MglA is required for the normal function of A-motility (adventurous), S-motility (social), fruiting body morphogenesis, and sporulation. MglA and its homologs differ from all eukaryotic and other prokaryotic GTPases because they have a threonine (Thr78) in place of the highly conserved aspartate residue of the consensus PM3 (phosphate-magnesium binding) region. To identify residues critical for MglA function or potential protein interactions, and explore the function of Thr78, the phenotypes of 18 mglA mutants were characterized. Results Nine mutants, with mutations predicted to alter residues that bind the guanine base or coordinate magnesium, did not produce detectable MglA. As expected, these mutants were mot- dev- because MglA is essential for these processes. Of the remaining nine mutants, seven showed a wild-type distribution pattern for MglA but fell into two categories with regard to function. Five of the seven mutants exhibited mild phenotypes, but two mutants, T78D and P80A, abolished motility and development. The localization pattern of MglA was abolished in two mutants that were mot- spo- and dev-. These two mutants were predicted to alter surface residues at Asp52 and Thr54, which suggests that these residues are critical for proper localization and may define a protein interaction site. Improving the consensus match with Ras at Thr78 abolished function of MglA. Only the conservative serine substitution was tolerated at this position. Merodiploid constructs revealed that a subset of alleles, including mglAD52A, were dominant and also illustrated that changing the balance of MglA and its co-transcribed partner, MglB, affects A-motility. Conclusion Our results suggest that GTP binding is critical for stability of MglA because MglA does not accumulate in mutants that cannot bind GTP. The threonine in PM3 of MglA proteins represents a novel modification of the highly conserved GTPase consensus at this position. The requirement for a hydroxyl group at this position may indicate that MglA is subject to modification under certain conditions. Proper localization of MglA is critical for both motility and development and likely involves protein interactions mediated by residues Asp52 and Thr54.
Collapse
|
9
|
Terry Powers JL, Mace KE, Parfrey H, Lee SJ, Zhang G, Riches DWH. TNF receptor-1 (TNF-R1) ubiquitous scaffolding and signaling protein interacts with TNF-R1 and TRAF2 via an N-terminal docking interface. Biochemistry 2010; 49:7821-9. [PMID: 20704259 DOI: 10.1021/bi100726n] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
TNF receptor-1 (TNF-R1) signal transduction is mediated through the assembly of scaffolding proteins, adaptors, and kinases. TNF receptor ubiquitous scaffolding and signaling protein (TRUSS), a 90.1 kDa TNF-R1-associated scaffolding protein, also interacts with TRAF2 and IKK and contributes to TNF-alpha-induced nuclear factor-kappaB (NF-kappaB) and c-Jun-NH(2)-terminal kinase (JNK) activation. Little is known about the mechanism of interaction among TRUSS, TNF-R1, and TRAF2. To address this issue, we used deletional and site-directed mutagenesis approaches to systematically investigate (i) the regions of TRUSS that interact with TNF-R1 and TRAF2 and (ii) the ability of TRUSS to self-associate to form higher-order complexes. Here we show that sequences located in the N-terminal (residues 1-248) and central (residues 249-440) regions of TRUSS are required to form a docking interface that supports binding to both TNF-R1 and TRAF2. While the C-terminal region (residues 441-797) did not directly interact with TNF-R1 or TRAF2, sequences located in this region were capable of self-association. Collectively, these data suggest that (i) the interaction between TNF-R1 and TRAF2 requires sequences located in the entire N-terminal half (residues 1-440) of TRUSS, (ii) the binding interface for TNF-R1 is closely linked with the TRAF2 binding interface, and (iii) the assembly of homomeric TRUSS complexes may contribute to its role in TNF-R1 signaling.
Collapse
Affiliation(s)
- Jennifer L Terry Powers
- Program in Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado 80206, USA
| | | | | | | | | | | |
Collapse
|
10
|
c-Fos proteasomal degradation is activated by a default mechanism, and its regulation by NAD(P)H:quinone oxidoreductase 1 determines c-Fos serum response kinetics. Mol Cell Biol 2010; 30:3767-78. [PMID: 20498278 DOI: 10.1128/mcb.00899-09] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The short-lived proto-oncoprotein c-Fos is a component of the activator protein 1 (AP-1) transcription factor. A large region of c-Fos is intrinsically unstructured and susceptible to a recently characterized proteasomal ubiquitin-independent degradation (UID) pathway. UID is active by a default mechanism that is inhibited by NAD(P)H:quinone oxidoreductase 1 (NQO1), a 20S proteasome gatekeeper. Here, we show that NQO1 binds and induces robust c-Fos accumulation by blocking the UID pathway. c-Jun, a partner of c-Fos, also protects c-Fos from proteasomal degradation by default. Our findings suggest that NQO1 protects monomeric c-Fos from proteasomal UID, a function that is fulfilled later by c-Jun. We show that this process regulates c-Fos homeostasis (proteostasis) in response to serum stimulation, phosphorylation, nuclear translocation, and transcription activity. In addition, we show that NQO1 is important to ensure immediate c-Fos accumulation in response to serum, since a delayed response was observed under low NQO1 expression. These data suggest that in vivo, protein unstructured regions determine the kinetics and the homeostasis of regulatory proteins. Our data provide evidence for another layer of regulation of key regulatory proteins that functions at the level of protein degradation and is designed to ensure optimal formation of functional complexes such as AP-1.
Collapse
|
11
|
St-Arnaud R, Mandic V, Elchaarani B. FIAT, the factor-inhibiting ATF4-mediated transcription, also represses the transcriptional activity of the bZIP factor FRA-1. Ann N Y Acad Sci 2010; 1192:338-43. [PMID: 20392257 DOI: 10.1111/j.1749-6632.2009.05211.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
FIAT is a leucine zipper protein whose name was coined for its interaction with ATF4 and subsequent blockage of ATF4-directed osteocalcin gene transcription. FIAT lacks a basic DNA-binding domain but contains three leucine zippers; it heterodimerizes with ATF4 to prohibit binding to DNA. FIAT could also potentially interact with additional basic domain-leucine zipper transcriptional regulators of osteoblast activity, such as FRA-1. We have found that FIAT inhibits transcriptional activation by a FRA-1/c-JUN heterodimer without affecting transcription mediated by a c-JUN homodimer. The repressor effect of FIAT on FRA-1-dependent transcription was measured using reporter constructs for the natural FRA-1 targets, Mmp-9 and Mgp. The FIAT-FRA-1 interaction is mediated through the second leucine zipper of FIAT. These data confirm an additional target of the FIAT transcriptional repressor activity and suggest that FIAT can both modulate early osteoblast activity by interacting with ATF4 and regulate later osteoblast function through inhibition of FRA-1.
Collapse
Affiliation(s)
- René St-Arnaud
- Shriners Hospital for Children, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
12
|
Mason JM. Electrostatic contacts in the activator protein-1 coiled coil enhance stability predominantly by decreasing the unfolding rate. FEBS J 2010; 276:7305-18. [PMID: 20050182 DOI: 10.1111/j.1742-4658.2009.07440.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The hypothesis is tested that Jun-Fos activator protein-1 coiled coil interactions are dominated during late folding events by the formation of intricate intermolecular electrostatic contacts. A previously derived cJun-FosW was used as a template as it is a highly stable relative of the wild-type cJun-cFos coiled coil protein (thermal melting temperature = 63 degrees C versus 16 degrees C), allowing kinetic folding data to be readily extracted. An electrostatic mutant, cJun(R)-FosW(E), was created to generate six Arg-Glu interactions at e-g'+1 positions between cJun(R) and FosW(E), and investigations into how their contribution to stability is manifested in the folding pathway were undertaken. The evidence now strongly indicates that the formation of interhelical electrostatic contacts exert their effect predominantly on the coiled coil unfolding/dissociation rate. This has major implications for future antagonist design whereby kinetic rules could be applied to increase the residency time of the antagonist-peptide complex, and therefore significantly increase the efficacy of the antagonist.
Collapse
Affiliation(s)
- Jody M Mason
- Department of Biological Sciences, University of Essex, Colchester, Essex, UK.
| |
Collapse
|
13
|
Malnou CE, Brockly F, Favard C, Moquet-Torcy G, Piechaczyk M, Jariel-Encontre I. Heterodimerization with different Jun proteins controls c-Fos intranuclear dynamics and distribution. J Biol Chem 2010; 285:6552-62. [PMID: 20053986 DOI: 10.1074/jbc.m109.032680] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The c-Fos proto-oncogenic transcription factor defines a multigene family controlling many processes both at the cell and the whole organism level. To bind to its target AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element DNA sequences in gene promoters and exert its transcriptional part, c-Fos must heterodimerize with other bZip proteins, its best studied partners being the Jun proteins (c-Jun, JunB, and JunD). c-Fos expression is regulated at many transcriptional and post-transcriptional levels, yet little is known on how its localization is dynamically regulated in the cell. Here we have investigated its intranuclear mobility using fluorescence recovery after photobleaching, genetic, and biochemical approaches. Whereas monomeric c-Fos is highly mobile and distributed evenly with nucleolar exclusion in the nucleus, heterodimerization with c-Jun entails intranuclear redistribution and dramatic reduction in mobility of c-Fos caused by predominant association with the nuclear matrix independently of any binding to AP-1/12-O-tetradecanoylphorbol-13-acetate-responsive element or cAMP-responsive element sequences. In contrast to c-Jun, dimerization with JunB does not detectably affect c-Fos mobility. However, dimerization with JunB affects intranuclear distribution with significant differences in the localization of c-Fos.c-Jun and c-Fos.JunB dimers. Moreover, c-Jun and JunB exert comparable effects on another Fos family member, Fra-1. Thus, we report a novel regulation, i.e. differentially regulated intranuclear mobility and distribution of Fos proteins by their Jun partners, and suggest the existence of intranuclear storage sites for latent c-Fos.c-Jun AP-1 complexes. This may affect the numerous physiopathological functions these transcription factors control.
Collapse
Affiliation(s)
- Cécile E Malnou
- Institut de Génétique Moléculaire de Montpellier, UMR5535, CNRS, 1919 route de Mende, 34293 Montpellier Cedex 5, France
| | | | | | | | | | | |
Collapse
|
14
|
Yu VWC, Gauthier C, St-Arnaud R. FIAT represses bone matrix mineralization by interacting with ATF4 through its second leucine zipper. J Cell Biochem 2008; 105:859-65. [PMID: 18680144 DOI: 10.1002/jcb.21881] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We have characterized FIAT, a 66 kDa leucine zipper (LZ) protein that dimerizes with activating transcription factor 4 (ATF4) to form inactive dimers that cannot bind DNA. Computer analysis identifies three putative LZ motifs within the FIAT amino acid sequence. We have used deletion- and/or site-specific mutagenesis to individually inactivate these motifs in order to identify the functional LZ that mediates the FIAT-ATF4 interaction. Amino acids 194-222 that encode the FIAT LZ2 were deleted (mutant FIAT ZIP2 DEL). We inactivated each zipper individually by replacing two or three leucine residues within each zipper by alanine residues. The engineered mutations were L142A/L149A (mutant M1, first zipper), L208A/L215A/L222A (mutant M2, second zipper), and L441A/L448A (mutant M3, third zipper). MC3T3-E1 osteoblastic cells with an integrated 1.3 kb mouse osteocalcin gene promoter fragment driving expression of luciferase were transfected with expression vectors for ATF4 and the various FIAT deletion- or site-specific mutants. Inhibition of ATF4-mediated transcription was compared between wild-type (WT) and LZ FIAT mutants. The deletion mutant FIAT ZIP2 DEL and the sequence-specific M2 mutant did not interact with ATF4 and were unable to inhibit ATF4-mediated transcription. The M1 or M3 mutations did not affect the ability of FIAT to contact ATF4 or to inhibit its transcriptional activity. Stable expression of WT FIAT in osteoblastic cells inhibited mineralization, but not expression of the FIAT ZIP2 DEL and M2 mutants. This structure-function analysis reveals that FIAT interacts with ATF4 and modulates its activity through its second leucine zipper motif.
Collapse
Affiliation(s)
- Vionnie W C Yu
- Genetics Unit, Shriners Hospital for Children, Montreal, Quebec, Canada
| | | | | |
Collapse
|
15
|
Thibodeaux GN, Cowmeadow R, Umeda A, Zhang Z. A tetracycline repressor-based mammalian two-hybrid system to detect protein-protein interactions in vivo. Anal Biochem 2008; 386:129-31. [PMID: 19111517 DOI: 10.1016/j.ab.2008.11.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 11/26/2008] [Indexed: 11/20/2022]
Abstract
A mammalian two-hybrid system (termed as trM2H) was developed to detect protein-protein interactions in vivo, based on the reconstitution of the functions the of tetracycline repressor (TetR). The system is sensitive enough to detect protein-protein interactions with K(d) up to 55microM in mammalian cells, and the system can be regulated by small molecules. This system can be used as an efficient genetic selection system to map protein-protein interactions in mammalian cells.
Collapse
|
16
|
Malnou CE, Salem T, Brockly F, Wodrich H, Piechaczyk M, Jariel-Encontre I. Heterodimerization with Jun family members regulates c-Fos nucleocytoplasmic traffic. J Biol Chem 2007; 282:31046-59. [PMID: 17681951 DOI: 10.1074/jbc.m702833200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
c-Fos proto-oncoprotein forms AP-1 transcription complexes with heterodimerization partners such as c-Jun, JunB, and JunD. Thereby, it controls essential cell functions and exerts tumorigenic actions. The dynamics of c-Fos intracellular distribution is poorly understood. Hence, we have combined genetic, cell biology, and microscopic approaches to investigate this issue. In addition to a previously characterized basic nuclear localization signal (NLS) located within the central DNA-binding domain, we identified a second NLS within the c-Fos N-terminal region. This NLS is non-classic and its activity depends on transportin 1 in vivo. Under conditions of prominent nuclear localization, c-Fos can undergo nucleocytoplasmic shuttling through an active Crm-1 exportin-independent mechanism. Dimerization with the Jun proteins inhibits c-Fos nuclear exit. The strongest effect is observed with c-Jun probably in accordance with the relative stabilities of the different c-Fos:Jun dimers. Retrotransport inhibition is not caused by binding of dimers to DNA and, therefore, is not induced by indirect effects linked to activation of c-Fos target genes. Monomeric, but not dimeric, Jun proteins also shuttle actively. Thus, our work unveils a novel regulation operating on AP-1 by demonstrating that dimerization is crucial, not only for active transcription complex formation, but also for keeping them in the compartment where they exert their transcriptional function.
Collapse
Affiliation(s)
- Cécile E Malnou
- Institut de Génétique Moléculaire de Montpellier, CNRS, UMR5535, 1919 Route de Mende, Montpellier F-34293, France
| | | | | | | | | | | |
Collapse
|
17
|
Jurado J, Fuentes-Almagro CA, Prieto-Álamo MJ, Pueyo C. Alternative splicing of c-fos pre-mRNA: contribution of the rates of synthesis and degradation to the copy number of each transcript isoform and detection of a truncated c-Fos immunoreactive species. BMC Mol Biol 2007; 8:83. [PMID: 17888145 PMCID: PMC2098773 DOI: 10.1186/1471-2199-8-83] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Accepted: 09/21/2007] [Indexed: 12/30/2022] Open
Abstract
Background Alternative splicing is a widespread mechanism of gene expression regulation. Previous analyses based on conventional RT-PCR reported the presence of an unspliced c-fos transcript in several mammalian systems. Compared to the well-defined knowledge on the alternative splicing of fosB, the physiological relevance of the unspliced c-fos transcript in regulating c-fos expression remains largely unknown. This work aimed to investigate the functional significance of the alternative splicing c-fos pre-mRNA. Results A set of primers was designed to demonstrate that, whereas introns 1 and 2 are regularly spliced from primary c-fos transcript, intron 3 remains unspliced in part of total transcript molecules. Here, the two species are referred to as c-fos-2 (+ intron 3) and spliced c-fos (- intron 3) transcripts. Then, we used a quantitatively rigorous approach based on real-time PCR to provide, for the first time, the actual steady-state copy numbers of the two c-fos transcripts. We tested how the mouse-organ context and mouse-gestational age, the synthesis and turnover rates of the investigated transcripts, and the serum stimulation of quiescent cells modulate their absolute-expression profiles. Intron 3 generates an in-frame premature termination codon that predicts the synthesis of a truncated c-Fos protein. This prediction was evaluated by immunoaffinity chromatography purification of c-Fos proteins. Conclusion We demonstrate that: (i) The c-fos-2 transcript is ubiquitously synthesized either in vivo or in vitro, in amounts that are higher or similar to those of mRNAs coding for other Fos family members, like FosB, ΔFosB, Fra-1 or Fra-2. (ii) Intron 3 confers to c-fos-2 an outstanding destabilizing effect of about 6-fold. (iii) Major determinant of c-fos-2 steady-state levels in cultured cells is its remarkably high rate of synthesis. (iv) Rapid changes in the synthesis and/or degradation rates of both c-fos transcripts in serum-stimulated cells give rise to rapid and transient changes in their relative proportions. Taken as a whole, these findings suggest a co-ordinated fine-tune of the two c-fos transcript species, supporting the notion that the alternative processing of the precursor mRNA might be physiologically relevant. Moreover, we detected a c-Fos immunoreactive species corresponding in mobility to the predicted truncated variant.
Collapse
Affiliation(s)
- Juan Jurado
- Universidad de Córdoba, Departamento de Bioquímica y Biología Molecular, Campus Rabanales, Edificio Severo Ochoa, planta-2, 14071-Córdoba, Spain
| | - Carlos A Fuentes-Almagro
- Universidad de Córdoba, Departamento de Bioquímica y Biología Molecular, Campus Rabanales, Edificio Severo Ochoa, planta-2, 14071-Córdoba, Spain
| | - María J Prieto-Álamo
- Universidad de Córdoba, Departamento de Bioquímica y Biología Molecular, Campus Rabanales, Edificio Severo Ochoa, planta-2, 14071-Córdoba, Spain
| | - Carmen Pueyo
- Universidad de Córdoba, Departamento de Bioquímica y Biología Molecular, Campus Rabanales, Edificio Severo Ochoa, planta-2, 14071-Córdoba, Spain
| |
Collapse
|
18
|
Mason JM, Schmitz MA, Müller KM, Arndt KM. Semirational design of Jun-Fos coiled coils with increased affinity: Universal implications for leucine zipper prediction and design. Proc Natl Acad Sci U S A 2006; 103:8989-94. [PMID: 16754880 PMCID: PMC1482553 DOI: 10.1073/pnas.0509880103] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Activator protein-1 (AP-1) is a crucial transcription factor implicated in numerous cancers. For this reason, nine homologues of the AP-1 leucine zipper region have been characterized: Fos (c-Fos, FosB, Fra1, and Fra2), Jun (c-Jun, JunB, and JunD), and semirational library-designed winning peptides FosW and JunW. The latter two were designed to specifically target c-Fos or c-Jun. They have been identified by using protein-fragment complementation assays combined with growth competition. This assay removes nonspecific, unstable, and protease susceptible library members from the pool, leaving winners with excellent drug potential. Thermal melts of all 45 possible dimeric interactions have been surveyed, with the FosW-c-Jun complex displaying a melting temperature (T(m)) of 63 degrees C, compared to only 16 degrees C for wild-type c-Fos-c-Jun interaction. This impressive 70,000-fold K(D) decrease is largely due to optimized core packing, alpha-helical propensity, and electrostatics. Contrastingly, due to a poor c-Fos core, c-Fos-JunW dimerizes with lower affinity. However the T(m) far exceeds wild-type c-Fos-c-Jun and averaged JunW and c-Fos, indicating a preference over either homodimer. Finally, and with wider implications, we have compiled a method for predicting interaction of parallel, dimeric coiled coils, using our T(m) data as a training set, and applying it to 59 bZIP proteins previously reported. Our algorithm, unlike others to date, accounts for helix propensity, which is found to be integral in coiled coil stability. Indeed, in applying the algorithm to these 59(2) bZIP interactions, we were able to correctly identify 92% of all strong interactions and 92% of all noninteracting pairs.
Collapse
Affiliation(s)
- Jody M. Mason
- Institut für Biologie III, Universität Freiburg, Schänzlestrasse 1, D-79104 Freiburg, Germany
| | - Mark A. Schmitz
- Institut für Biologie III, Universität Freiburg, Schänzlestrasse 1, D-79104 Freiburg, Germany
| | - Kristian M. Müller
- Institut für Biologie III, Universität Freiburg, Schänzlestrasse 1, D-79104 Freiburg, Germany
| | - Katja M. Arndt
- Institut für Biologie III, Universität Freiburg, Schänzlestrasse 1, D-79104 Freiburg, Germany
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
19
|
Yu M, Sha H, Gao Y, Zeng H, Zhu M, Gao X. Alternative 3′ UTR polyadenylation of Bzw1 transcripts display differential translation efficiency and tissue-specific expression. Biochem Biophys Res Commun 2006; 345:479-85. [PMID: 16690031 DOI: 10.1016/j.bbrc.2006.04.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Accepted: 04/21/2006] [Indexed: 11/23/2022]
Abstract
BZW1 is a conserved regulatory factor for transcriptional control of histone H4 gene at the G1/S transition. In this study, three Bzw1 transcripts were identified in mice with two long forms (approximately 2.9 kb) expressed ubiquitously at low level, and a short transcript of 1.8 kb expressed at high level exclusively in testis. These different transcripts share the same 5' UTR and coding sequence, but differ in the length of 3' UTR by utilizing alternative polyadenylation sites. Different translation efficiencies were observed in the cells transfected with chimeric EGFP-Bzw1 genes tailed with different 3' UTRs. Our results demonstrate that Bzw1 transcripts are alternatively polyadenylated and expressed in tissue-specific pattern.
Collapse
Affiliation(s)
- Mingyan Yu
- Model Animal Research Center and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China
| | | | | | | | | | | |
Collapse
|
20
|
Kuramoto N, Kubo K, Ogita K, Pláteník J, Balcar VJ, Takarada T, Nakamichi N, Yoneda Y. Nuclear condensation of cyclic adenosine monophosphate responsive element-binding protein in discrete murine brain structures. J Neurosci Res 2005; 80:667-76. [PMID: 15880467 DOI: 10.1002/jnr.20504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We have directed a polyclonal antibody against an oligo-peptide (123-136) of the transcription factor cyclic AMP responsive element-binding protein (CREB) including the serine residue at 133. Rabbit sera were purified by ammonium sulfate precipitation, followed by affinity chromatography to homogeneity on one-dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis. The purified antibody not only induced marked supershift of CREB binding, without affecting binding of activator protein-1 on gel retardation electrophoresis, but also differentiated between CREB and CREB phosphorylated at serine133 in brain nuclear fractions on Western blotting. Immunoreactive CREB was detected in both cytosolic and nuclear fractions of discrete murine brain structures but was more highly condensed in cerebellum than in neocortex and hippocampus. Incubation of brain nuclear fractions led to a marked export of immunoreactive CREB in a temperature-dependent manner, whereas the temperature-dependent export activity was significantly lower in cerebellum than in other brain structures. Suppression of general new protein synthesis by cycloheximide (500 mg/kg, i.p.) in vivo resulted in a significant decrease in the nuclear CREB level, with a concomitant increase in the cytosolic level in hippocampus, but not in cerebellum. These results suggest that the nuclear export activity might vary from region to region in murine brains through a hitherto unidentified mechanism other than the nuclear localization signal, to result in different nuclear condensation ratios for subsequent elicitation of differential transcriptional activities by the constitutive transcription factor CREB in the nucleus.
Collapse
Affiliation(s)
- Nobuyuki Kuramoto
- Laboratory of Molecular Pharmacology, Division of Pharmaceutical Sciences, Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Ishikawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Segel GB, Woodlock TJ, Xu J, Li L, Felgar RE, Ryan DH, Lichtman MA, Wang N. Early gene activation in chronic leukemic B lymphocytes induced toward a plasma cell phenotype. Blood Cells Mol Dis 2003; 30:277-87. [PMID: 12737946 DOI: 10.1016/s1079-9796(03)00035-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of lymphocytes that are arrested at an intermediate stage of B lymphocyte development. CLL B lymphocytes transform (mature) to a plasmacytic phenotype with loss of CD19 and CD20 and the appearance of cytoplasmic immunoglobulin when treated in vitro with phorbol esters. We have used array hybridization technology to describe gene expression patterns for untreated and tetradecanoyl phorbol acetate (TPA)-treated CLL B cells at 5, 10, and 20 min following initial TPA exposure. Three genes, early growth response factor 1 (EGR-1), dual specificity phosphatase 2, and CD69 (early T-cell activation antigen), showed a 2.0-fold or greater increase in mRNA transcription at four or more of six time points in two studies. Upregulation of expression of these genes was confirmed by real-time polymerase chain reaction in the TPA-treated cells of four CLL patients. A progressive increase in gene expression was observed during the 20-min time course for all three genes. In addition, protein expression of EGR-1 and CD69 was increased as measured by immunofluorescence cell analysis. Several genes (PKC, n-myc, jun D, and BCL-2) previously reported as overexpressed in CLL lymphocytes were overexpressed in these studies also, but were not altered by TPA treatment. Genes for proteins whose upregulation requires hours of TPA exposure (the 4F2hc component of the L-system amino acid transporter, prohibition, and hsp60) were assessed, and their later expression contrasted with the early expression of EGR-1, dual specificity phosphatase 2, and CD69. EGR-1 encodes a zinc-finger transcription factor that is induced by pokeweed mitogen and TPA and promotes B lymphocyte maturation. The dual specificity phosphatase 2 encodes an enzyme that reverses mitogen activated protein kinase cell activation by dephosphorylation. The CD69 protein is induced by TPA in thymocytes and is a type II transmembrane signaling molecule in hematopoietic cells. These findings suggest that the products of these three genes may be central to early steps in the TPA-induced evolution of CLL B cells to a plasmacytic phenotype.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, Differentiation, T-Lymphocyte/genetics
- B-Lymphocytes/drug effects
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- DNA-Binding Proteins/genetics
- Early Growth Response Protein 1
- Gene Expression Profiling
- Humans
- Immediate-Early Proteins/genetics
- Lectins, C-Type
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Oligonucleotide Array Sequence Analysis
- Plasma Cells/cytology
- Protein Phosphatase 2
- Protein Tyrosine Phosphatases/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Tetradecanoylphorbol Acetate/pharmacology
- Time Factors
- Transcription Factors/genetics
- Transcription, Genetic
Collapse
Affiliation(s)
- George B Segel
- Department of Pediatrics, and Unity Health System, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Ferrara P, Andermarcher E, Bossis G, Acquaviva C, Brockly F, Jariel-Encontre I, Piechaczyk M. The structural determinants responsible for c-Fos protein proteasomal degradation differ according to the conditions of expression. Oncogene 2003; 22:1461-74. [PMID: 12629509 DOI: 10.1038/sj.onc.1206266] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
c-fos gene is expressed constitutively in a number of tissues as well as in certain tumor cells and is inducible, in general rapidly and transiently, in virtually all other cell types by a variety of stimuli. Its protein product, c-Fos, is a short-lived transcription factor that heterodimerizes with various protein partners within the AP-1 transcription complex via leucine zipper/leucine zipper interactions for binding to specific DNA sequences. It is mostly, if not exclusively, degraded by the proteasome. To localize the determinant(s) responsible for its instability, we have conducted a genetic analysis in which the half-lives of c-Fos mutants and chimeras made with the stable EGFP reporter protein were compared under two experimental conditions taken as example of continous and inducible expression. Those were constitutive expression in asynchronously growing Balb/C 3T3 mouse embryo fibroblasts and transient induction in the same cells undergoing the G0/G1 phase transition upon stimulation by serum. Our work shows that c-Fos is degraded faster in synchronous- than in asynchronous cells. This difference in turnover is primarily accounted for by several mechanisms. First, in asynchronous cells, a unique C-terminal destabilizer is active whereas, in serum-stimulated cells two destabilizers located at both extremities of the protein are functional. Second, heterodimerization and/or binding to DNA accelerates protein degradation only during the G0/G1 phase transition. Adding another level of complexity to turnover control, phosphorylation at serines 362 and 374, which are c-Fos phosphorylation sites largely modified during the G0/G1 phase transition, stabilizes c-Fos much more efficiently in asynchronous than in serum-stimulated cells. In both cases, the reduced degradation rate is due to inhibition of the activity of the C-terminal destabilizer. However, in serum-stimulated cells, this effect is partially masked by the activation of the N-terminal destabilizer and basic domain/leucine zipper-dependent mechanisms. Taken together, our data show that multiple degradation mechanisms, differing according to the conditions of expression, may operate on c-Fos to ensure a proper level and/or timing of expression. Moreover, they also indicate that the half-life of c-Fos during the G0/G1 phase transition is determined by a delicate balance between opposing stabilizing and destabilizing mechanisms operating at the same time.
Collapse
|
23
|
Localization of activator protein-1 complex with DNA binding activity in mitochondria of murine brain after in vivo treatment with kainate. J Neurosci 2002. [PMID: 11923421 DOI: 10.1523/jneurosci.22-07-02561.2002] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To elucidate mechanisms underlying mitochondrial dysfunctions induced by glutamate, we have examined the effects of in vivo treatment with the ionotropic glutamate receptor agonist kainate on localization of the transcription factor activator protein-1 (AP-1) in mitochondria as well as nuclei of murine brain. A systemic administration of kainate dramatically enhanced AP-1 DNA binding in both mitochondrial and nuclear extracts of mouse cerebral cortex and hippocampus 1 hr to 3 d later. Unlabeled AP-1 probe selectively competed for AP-1 DNA binding in mitochondrial extracts of cortex and hippocampus obtained from mice injected with kainate. Supershift and immunoblotting analyses revealed participation of c-Fos, Fos-B, and Jun-B proteins in potentiation by kainate of mitochondrial AP-1 DNA binding in cortex and hippocampus. An immunohistochemical study demonstrated marked expression by kainate of c-Fos protein in the pyramidal and dentate granular layers, whereas an immunoelectron microscopic analysis showed localization of c-Fos protein within mitochondria, as well as nuclei, of the CA1 pyramidal and dentate granular cells in hippocampus obtained 2 hr after the administration of kainate. Mitochondrial AP-1 DNA binding was inhibited by particular unlabeled oligonucleotides containing sequences similar to the AP-1 site found in the noncoding region of mitochondrial DNA. Kainate markedly potentiated binding of radiolabeled oligonucleotide probes containing sequences effective in competing for AP-1 DNA binding in hippocampal mitochondrial extracts. These results suggest that kainate may facilitate expression of the AP-1 complex and subsequent translocation into mitochondria to participate in mechanisms associated with transcriptional regulation of mitochondrial DNA in murine hippocampus.
Collapse
|
24
|
Mitsuno Y, Maeda S, Yoshida H, Hirata Y, Ogura K, Akanuma M, Kawabe T, Shiratori Y, Omata M. Helicobacter pylori activates the proto-oncogene c-fos through SRE transactivation. Biochem Biophys Res Commun 2002; 291:868-74. [PMID: 11866445 DOI: 10.1006/bbrc.2002.6530] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Epidemiological studies have demonstrated a strong association between Helicobacter pylori infection and gastric cancer. However, there have been few detailed studies on the mechanism of cellular proliferation by H. pylori. Thus, we examined activation of the proto-oncogene c-fos to elucidate the underlying mechanism of cell proliferation caused by H. pylori. Activation of c-fos was evaluated in human gastric cancer cells (TMK1) by Northern blot and reporter assays with deletion analysis of the c-fos transcriptional control region. c-fos promoter activation and transcription were enhanced when cocultured with cag-positive strains. H. pylori-mediated c-fos promoter activation was inhibited by MEK1/2 inhibitor (U0126). The deletion analysis indicated that serum response element (SRE) was required for the activation of c-fos by H. pylori. In conclusion, c-fos promoter activation and transcription were enhanced through the activation of extracellular signal-regulated kinases (ERK)/mitogen-activated protein kinase (MAPK) cascade in gastric cancer cells when cocultured with H. pylori possessing intact cag PAI. SRE is required for the activation of c-fos by H. pylori. These results suggest a direct involvement of H. pylori infection in cellular proliferation, which may play a role in neoplastic transformation.
Collapse
Affiliation(s)
- Yuzo Mitsuno
- Department of Gastroenterology, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mitchell ES, Keller RW, Snyder-Keller A. Immediate-early gene expression in concurrent prenatal ethanol- and/or cocaine-exposed rat pups: intrauterine differences in cocaine levels and Fos expression. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2002; 133:141-9. [PMID: 11882344 DOI: 10.1016/s0165-3806(02)00282-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Concurrent use of cocaine and ethanol is a common mode of abuse. Cocaine and ethanol have distinctive pharmacologies but both have been shown to cause uterine vasoconstriction and fetal hypoxia. We developed a paradigm of chronic ethanol exposure via liquid diet coupled with binge cocaine exposure on the last day of gestation. Lipton et al. demonstrated unequal segregation of cocaine in rat fetuses as a function of proximal-distal location in the uterus, indicating a differential vasoconstriction of the two main arteries supplying the uterus in rats receiving cocaine. By performing C-sections after exposure to cocaine, we were able to measure the cocaine content and immediate-early gene (IEG) induction in the brains of fetuses according to their intrauterine position and assess the potentially vasoconstrictive effect of ethanol. HPLC analysis of fetal brains exposed to cocaine supported the study of Lipton et al.: fetuses from the proximal (lower) end of the uterus had more cocaine than fetuses from the distal (upper) end. Concurrent ethanol decreased the amount of cocaine reaching the fetuses and diminished the proximal-distal gradient. There were increased numbers of Fos-immunoreactive cells in fetuses exposed to both ethanol and cocaine compared to cocaine binge only. Additionally, the gradient of c-fos induction observed as a function of intrauterine position in cocaine-treated rats was in the opposite direction: most distal fetuses generally had the most Fos-immunoreactive cells. These results indicate that IEG induction in fetal brains exposed to cocaine and ethanol may be more related to hypoxic consequences of prenatal drug exposure.
Collapse
Affiliation(s)
- Ellen S Mitchell
- Department of Biomedical Sciences, State University at Albany, Albany, NY, USA.
| | | | | |
Collapse
|
26
|
Fedida D, Hesketh JC. Gating of voltage-dependent potassium channels. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2001; 75:165-99. [PMID: 11376798 DOI: 10.1016/s0079-6107(01)00006-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of voltage-dependent ion channels is primarily controlled by the applied potential difference across the membrane. For potassium channels the Drosophila Shaker channel has served as an archetype of all other potassium channels in studies of activation mechanisms. In the Shaker potassium channel much of the voltage sensitivity is conferred by the S4 transmembrane helix, which contains seven positively charged residues. During gating, the movement of these charges produces gating currents. Mutagenic and fluorescence studies indicate that four of these residues are particularly important and contribute to the majority of gating charge, R362, R365, R368 and R371. The channel is thought to dwell in several closed states prior to opening. Ionic-charge pairing with negatively charged residues in the S2 and S3 helices is thought to be important in regulating these closed states and detailed kinetic models have attempted to define the kinetics and charge of the transitions between these states. Neutral residues throughout the S4 and S5 helices are thought to control late steps in channel opening and may have important roles in modulating the stability of the open state and late closed states. In response to depolarization, the S4 helix is thought to undergo a rotational translation and this movement is also important in studies of the movement of the pore helices, S5 and S6, during opening. This review will examine residues that are important during activation as well as kinetic models that have attempted to quantitatively define the activation pathway of voltage-dependent potassium channels.
Collapse
Affiliation(s)
- D Fedida
- Department of Physiology, University of British Columbia, 2146 Health Sciences Mall, V6T 1Z3, Vancouver, Canada.
| | | |
Collapse
|
27
|
Hess J, Porte D, Munz C, Angel P. AP-1 and Cbfa/runt physically interact and regulate parathyroid hormone-dependent MMP13 expression in osteoblasts through a new osteoblast-specific element 2/AP-1 composite element. J Biol Chem 2001; 276:20029-38. [PMID: 11274169 DOI: 10.1074/jbc.m010601200] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The expression of MMP13 (collagenase-3), a member of the matrix metalloproteinase family, is increased in vivo as well as in cultured osteosarcoma cell lines by parathyroid hormone (PTH), a major regulator of calcium homeostasis. Binding sites for AP-1 and Cbfa/Runt transcription factors in close proximity have been identified as cis-acting elements in the murine and rat mmp13 promoter required for PTH-induced expression. The cooperative function of these factors in response to PTH in osteoblastic cells suggests a direct interaction between AP-1 and Cbfa/Runt transcription factors. Here, we demonstrate interaction between c-Jun and c-Fos with Cbfa/Runt proteins. This interaction depends on the leucine zipper of c-Jun or c-Fos and the Runt domain of Cbfa/Runt proteins, respectively. Moreover, c-Fos interacts with the C-terminal part of Cbfa1 and Cbfa2, sharing a conserved transcriptional repression domain. In addition to the distal osteoblast-specific element 2 (OSE2) element in the murine and rat mmp13 promoter, we identified a new proximal OSE2 site overlapping with the TRE motif. Both interaction of Cbfa/Runt proteins with AP-1 and the presence of a functional proximal OSE2 site are required for enhanced transcriptional activity of the mmp13 promoter in transient transfected fibroblasts and in PTH-treated osteosarcoma cells.
Collapse
Affiliation(s)
- J Hess
- Deutsches Krebsforschungszentrum Heidelberg, Division of Signal Transduction and Growth Control (B0800), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
28
|
Jérôme V, Müller R. A synthetic leucine zipper-based dimerization system for combining multiple promoter specificities. Gene Ther 2001; 8:725-9. [PMID: 11406767 DOI: 10.1038/sj.gt.3301443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2000] [Accepted: 01/30/2001] [Indexed: 11/09/2022]
Abstract
One of the biggest challenges facing gene therapy is the development of vectors that direct the activity of therapeutic genes specifically to the sites of disease. To achieve this goal, the restriction of transgene transcription via synthetic promoters that are endowed with multiple specificities represents a particularly promising strategy. Towards this end, we have developed a generally applicable strategy (DCTF system) where a synthetic promoter is driven by an artificial heterodimeric transcription factor whose DNA-binding and transactivating subunits are expressed from two promoters with different selectivity. A crucial determinant of the DCTF system is the heterodimerization interface that should provide for a high affinity interaction without interference by endogenous proteins. Here, we describe such a dimerization system based on engineered Fos and Jun leucine zippers. We show the usefulness of this system for the combination of cell type-specific and cell cycle-regulated transcription and demonstrate its functionality in an in vivo setting.
Collapse
Affiliation(s)
- V Jérôme
- Institute of Molecular Biology and Tumor Research (IMT), Philipps University, Emil-Mannkopff-Strasse 2, 35033 Marburg, Germany
| | | |
Collapse
|
29
|
Abstract
Genetically modified mice have provided important insights into the biological functions of the dimeric transcription factor complex AP-1. Extensive analyses of mice and cells with genetically modified Fos or Jun proteins provide novel insights into the physiological functions of AP-1 proteins. Using knock-out strategies it was found that some components, such as c-Fos, FosB and JunD are dispensable, whereas others, like c-Jun, JunB and Fra-1 are essential in embryonic development and/or in the adult organism. Besides the specific roles of AP-1 proteins in developmental processes, we are beginning to obtain a better molecular understanding of the cell-context dependent function of AP-1 in cell proliferation and apoptosis, in bone biology as well as in multistep tumorigenesis.
Collapse
Affiliation(s)
- W Jochum
- Research Institute of Molecular Pathology (I.M.P.), Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | |
Collapse
|
30
|
Abstract
The AP-1 transcription factor is composed of a mixture of homo- and hetero-dimers formed between Jun and Fos proteins. The different Jun and Fos family members vary significantly in their relative abundance and their interactions with additional proteins generating a complex network of transcriptional regulators. Thus, the functional activity of AP-1 in any given cell depends on the relative amount of specific Jun/Fos proteins which are expressed, as well as other potential interacting proteins. This diversity of AP-1 components has complicated our understanding of AP-1 function and resulted in a paucity of information about the precise role of individual AP-1 members in distinct cellular processes. We shall discuss recent studies which suggest that different Jun and Fos family members may have both opposite and overlapping functions in cellular proliferation and cell fate.
Collapse
Affiliation(s)
- F Mechta-Grigoriou
- Unité des virus oncogenes, CNRS URA 1644, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | |
Collapse
|
31
|
Jaya P, Premkumar M, Varman Thampan R. Free leucine dissociates homo- and heterodimers formed between proteins containing leucine heptad repeats. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1499:171-9. [PMID: 11341964 DOI: 10.1016/s0167-4889(00)00132-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A highly specific method for the dissociation of protein dimers has been developed. The method involves exposure of the dimers to free leucine at a concentration ranging between 3 and 10 mM. Using this method it has been possible to dissociate goat uterine oestrogen receptor homodimers, heterodimers formed between the non-activated oestrogen receptor (naER) and the oestrogen receptor activation factor (E-RAF) of the goat uterus, c-jun homodimers derived from bovine bone marrow and also glucocorticoid receptor homodimers isolated from rat liver cytosol. The pattern of dimer dissociation by leucine clearly differentiates two classes of proteins. The first is represented by steroid hormone receptors where dimerization is apparently contributed by both coiled-coil dimerization interfaces and the conserved heptad repeats of leucine. The second is represented by oncoproteins like c-fos and c-jun which dimerize through the exclusive involvement of leucine zippers. The patterns of dissociation of these two groups of proteins from the concerned affinity columns are distinctly different. This indicates a possibility that the elution pattern may be used as a yardstick to determine whether two proteins dimerize through the exclusive involvement of leucine zippers or whether coiled-coil interfaces are also involved in the dimerization process.
Collapse
Affiliation(s)
- P Jaya
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Thiruvananthapuram 695 014, Kerala, India
| | | | | |
Collapse
|
32
|
Cheng Y, Zhizhin I, Perlman RL, Mangoura D. Prolactin-induced cell proliferation in PC12 cells depends on JNK but not ERK activation. J Biol Chem 2000; 275:23326-32. [PMID: 10807911 DOI: 10.1074/jbc.m001837200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The effects of pituitary and extrapituitary prolactin include cellular proliferation and differentiation. PC12 cells was used as a model to delineate respective signaling of prolactin. Prolactin acted as a mitogen for undifferentiated PC12 cells, as measured by significant increases in bromodeoxyuridine incorporation and in cell numbers, with an efficacy equal to epidermal growth factor. Both the long and short form of the prolactin receptor was expressed, yet only the long isoform was tyrosine-phosphorylated upon agonist binding. Functional prolactin receptor signaling was further demonstrated in the activation of JAK2 and phosphorylation activation of the transcription factors Stat1, -3, and -5a. Surprisingly, prolactin stimulated a sustained activation of Raf-B, without activation of the MAP kinases ERK1 or -2. Instead, in solid phase kinase assays using a glutathione S-transferase-c-Jun fusion protein (amino acids 1-79) as the substrate, a significant activation of the mitogen-activated protein Janus kinase (c-Jun N-terminal kinase; JNK) was observed. The prolactin-induced activation of JNK was prolonged and accompanied by a significant increase in c-Jun mRNA abundance and c-Jun protein synthesis. Moreover, analysis of bromodeoxyuridine incorporation at the single cell level revealed that epidermal growth factor-dependent incorporation was inhibited by PD98059 and independent of SB203580, whereas prolactin-induced incorporation was ERK and mitogen-activated protein kinase p38 independent but was abolished with JNK inhibition by 30 microm SB203580. Our studies suggest that prolactin may have a role in the growth of PC12 cells, where it stimulates concurrent mitogenic and differentiation-promoting signaling pathways.
Collapse
Affiliation(s)
- Y Cheng
- Kennedy Center, Department of Pediatrics, Committee on Neurobiology and Committee Cell Physiology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
33
|
Meyer-ter-Vehn T, Covacci A, Kist M, Pahl HL. Helicobacter pylori activates mitogen-activated protein kinase cascades and induces expression of the proto-oncogenes c-fos and c-jun. J Biol Chem 2000; 275:16064-72. [PMID: 10747974 DOI: 10.1074/jbc.m000959200] [Citation(s) in RCA: 192] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori is an etiological agent in the development of mucosa-associated lymphoid tissue lymphoma and gastric adenocarcinoma. Patients infected with H. pylori carry a 3-6-fold increased risk of developing cancer compared with uninfected individuals. H. pylori strains expressing the cytotoxin-associated antigen A (CagA) are more frequently associated with the development of neoplasia than cagA-negative strains. However, the molecular mechanism by which H. pylori causes neoplastic transformation remains unclear. Here we report that exposure of gastric epithelial cells to H. pylori induces activation of the transcription factor activator protein 1. Activation of the proto-oncogenes c-fos and c-jun is strongly induced. We show that H. pylori activates the ERK/MAP kinase cascade, resulting in Elk-1 phosphorylation and increased c-fos transcription. H. pylori strains that do not express CagA or that are mutated in cag genes encoded by the CagI pathogenicity island do not induce activator protein 1, MAP kinase activity, or c-fos or c-jun activation. Proto-oncogene activation may represent a crucial step in the pathomechanism of H. pylori induced neoplasia.
Collapse
Affiliation(s)
- T Meyer-ter-Vehn
- Division of Experimental Anaesthesiology, University Hospital Freiburg, Center for Tumor Biology, P. O. Box 1120, 79106 Freiburg, Germany
| | | | | | | |
Collapse
|
34
|
Park JM, Bauer SB, Freeman MR, Peters CA. Oxybutynin chloride inhibits proliferation and suppresses gene expression in bladder smooth muscle cells. J Urol 1999; 162:1110-4. [PMID: 10458442 DOI: 10.1097/00005392-199909000-00048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE We test the hypothesis that oxybutynin chloride inhibits bladder smooth muscle cell proliferation. MATERIALS AND METHODS Cultured rat bladder smooth muscle cells were grown in Medium 199 supplemented with 10% fetal bovine serum in the presence of 0, 1, 10 and 100 microM. oxybutynin. Cell proliferation was assessed by counting cell numbers 48 and 96 hours after plating. To investigate the role of oxybutynin in bladder smooth muscle cell proliferation after mechanical stretch, cells were grown on silicone elastomer bottomed culture plates and subjected to cyclical stretch-relaxation for 48 hours in the presence of 10 microM. oxybutynin. Deoxyribonucleic acid synthesis was assessed by tritiated thymidine incorporation assay. To examine the effect of oxybutynin on stretch activated gene expression, bladder smooth muscle cells were subjected to stretch-relaxation for 2 hours with and without 10 microM. oxybutynin, and relative c-jun messenger (m) ribonucleic acid (RNA) levels were assessed by semiquantitative reverse transcriptase-polymerase chain reaction with normalization to glyceraldehyde-3-phosphate dehydrogenase mRNA levels. RESULTS The serum stimulated increase in bladder smooth muscle cell growth was inhibited by oxybutynin in a dose dependent manner. In bladder smooth muscle cells there was a 4.7-fold increase in deoxyribonucleic acid synthesis after mechanical stretch, which decreased by 40% (p <0.01) when cells were stretched in the presence of oxybutynin. Stretch stimulated significant increase in c-jun mNRA levels, which was significantly decreased by oxybutynin. CONCLUSIONS Oxybutynin chloride inhibits bladder smooth muscle cell proliferation induced by serum and mechanical stretch. A potential mechanism by which oxybutynin inhibits proliferation may be the down regulation of growth promoting genes, such as c-jun. We speculate that oxybutynin may be useful for preventing permanent hypertrophic bladder changes in addition to decreasing intravesical pressure.
Collapse
Affiliation(s)
- J M Park
- Department of Urology, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
35
|
Park JM, Bauer SB, Freeman MR, Peters CA. Oxybutynin chloride inhibits proliferation and suppresses gene expression in bladder smooth muscle cells. J Urol 1999; 162:1110-4. [PMID: 10458442 DOI: 10.1016/s0022-5347(01)68086-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE We test the hypothesis that oxybutynin chloride inhibits bladder smooth muscle cell proliferation. MATERIALS AND METHODS Cultured rat bladder smooth muscle cells were grown in Medium 199 supplemented with 10% fetal bovine serum in the presence of 0, 1, 10 and 100 microM. oxybutynin. Cell proliferation was assessed by counting cell numbers 48 and 96 hours after plating. To investigate the role of oxybutynin in bladder smooth muscle cell proliferation after mechanical stretch, cells were grown on silicone elastomer bottomed culture plates and subjected to cyclical stretch-relaxation for 48 hours in the presence of 10 microM. oxybutynin. Deoxyribonucleic acid synthesis was assessed by tritiated thymidine incorporation assay. To examine the effect of oxybutynin on stretch activated gene expression, bladder smooth muscle cells were subjected to stretch-relaxation for 2 hours with and without 10 microM. oxybutynin, and relative c-jun messenger (m) ribonucleic acid (RNA) levels were assessed by semiquantitative reverse transcriptase-polymerase chain reaction with normalization to glyceraldehyde-3-phosphate dehydrogenase mRNA levels. RESULTS The serum stimulated increase in bladder smooth muscle cell growth was inhibited by oxybutynin in a dose dependent manner. In bladder smooth muscle cells there was a 4.7-fold increase in deoxyribonucleic acid synthesis after mechanical stretch, which decreased by 40% (p <0.01) when cells were stretched in the presence of oxybutynin. Stretch stimulated significant increase in c-jun mNRA levels, which was significantly decreased by oxybutynin. CONCLUSIONS Oxybutynin chloride inhibits bladder smooth muscle cell proliferation induced by serum and mechanical stretch. A potential mechanism by which oxybutynin inhibits proliferation may be the down regulation of growth promoting genes, such as c-jun. We speculate that oxybutynin may be useful for preventing permanent hypertrophic bladder changes in addition to decreasing intravesical pressure.
Collapse
Affiliation(s)
- J M Park
- Department of Urology, Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
36
|
Azuma Y, Ogita K, Yoneda Y. Constitutive expression of cytoplasmic activator protein-1 with DNA binding activity and responsiveness to ionotropic glutamate signals in the murine hippocampus. Neuroscience 1999; 92:1295-308. [PMID: 10426485 DOI: 10.1016/s0306-4522(99)00090-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Gel retardation electrophoresis revealed that cytosolic fractions contained DNA binding activity of the transcription factor activator protein-1 with profiles different from those reported in nuclear extracts in murine brain. In particular, activator protein-1 DNA binding was almost undetectable at 25 degrees C in the presence of both KCl and MgCl2 in cytosol fractions. Moreover, cytoplasmic activator protein-1 binding occurred at three different mobilities on the gel when determined at 2 degrees C in the absence of MgCl2. Systemic administration of N-methyl-D-aspartate and kainate led to marked potentiation of cytoplasmic activator protein-1 binding detected as slow bands in the murine hippocampus, without markedly affecting that as a fast band. Immunoblotting and supershift assays revealed much higher expression of both immunoreactive c-Jun and c-Fos in hippocampal cytosolic fractions in response to the administration of kainate than N-methyl-D-aspartate. These results suggest that activator protein-1 may be constitutively expressed in the cytoplasm with DNA binding activity and responsiveness to ionotropic glutamate signals in a manner different from that in the nucleus in the murine hippocampus.
Collapse
Affiliation(s)
- Y Azuma
- Department of Pharmacology, Setsunan University, Hirakata, Osaka, Japan
| | | | | |
Collapse
|
37
|
Abstract
The Fos and Jun oncoproteins form dimeric complexes that stimulate transcription of genes containing activator protein-1 regulatory elements. We found, by representational difference analysis, that expression of DNA 5-methylcytosine transferase (dnmt1) in fos-transformed cells is three times the expression in normal fibroblasts and that fos-transformed cells contain about 20 percent more 5-methylcytosine than normal fibroblasts. Transfection of the gene encoding Dnmt1 induced morphological transformation, whereas inhibition of dnmt1 expression or activity resulted in reversion of fos transformation. Inhibition of histone deacetylase, which associates with methylated DNA, also caused reversion. These results suggest that fos may transform cells through alterations in DNA methylation and in histone deacetylation.
Collapse
Affiliation(s)
- A V Bakin
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
38
|
|
39
|
Jang M, Pezzuto JM. Effects of resveratrol on 12-O-tetradecanoylphorbol-13-acetate-induced oxidative events and gene expression in mouse skin. Cancer Lett 1998; 134:81-9. [PMID: 10381133 DOI: 10.1016/s0304-3835(98)00250-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Resveratrol (3,5,4'-trihydroxy-trans-stilbene) is a natural product shown to inhibit carcinogen-induced pre-neoplastic lesions in mouse mammary organ culture and 12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted mouse skin tumors. Application of TPA to mouse skin induces oxidative stress, as evidenced by numerous biochemical responses, including significant generation of H2O2 and enhanced levels of myeloperoxidase and oxidized glutathione reductase activities and decreases in glutathione levels and superoxide dismutase activity. TPA treatment also elevates the expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), c-myc, c-fos, c-jun, transforming growth factor-beta1 (TGF-beta1) and tumor necrosis factor-alpha (TNF-alpha). As currently reported, pre-treatment of mouse skin with resveratrol negated several of these TPA-induced effects in a dose-dependent manner. H2O2 and glutathione levels were restored to control levels, as were myeloperoxidase, oxidized glutathione reductase and superoxide dismutase activities. As judged by reverse transcriptase-polymerase chain reaction (RT-PCR), TPA-induced increases in the expression of c-fos and TGF-beta1 were selectively inhibited. These data suggest that resveratrol inhibits tumorigenesis in mouse skin through interference with pathways of reactive oxidants and possibly by modulating the expression of c-fos and TGF-beta1.
Collapse
Affiliation(s)
- M Jang
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 60612, USA
| | | |
Collapse
|
40
|
Arrizubieta MJ, Bandman E. The role of interhelical ionic interactions in myosin rod assembly. Biochem Biophys Res Commun 1998; 244:588-93. [PMID: 9514858 DOI: 10.1006/bbrc.1998.8105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interhelical electrostatic interactions at specific heptad positions can regulate dimerization specificity of alpha-helical coiled-coils. We have analyzed 20 vertebrate myosin sequences from a variety of organisms and tissues in order to determine if interhelical ionic interactions correlate with the observed myosin dimerization specificity. We find that the sites for potential interhelical ion pairing are identical in virtually all sarcomeric myosins whether they form homo- or heterodimers. We also show that smooth muscle and non-muscle myosin rod sequences exhibit a different conserved pattern of potential interhelical ion pairing. These observations suggest that myosin rod residues involved in interhelical electrostatic interactions do not regulate dimerization specificity, but may contribute to the specific arrangements of myosin molecules that determine differences in the filament morphology of sarcomeric and non-sarcomeric muscles.
Collapse
Affiliation(s)
- M J Arrizubieta
- Department of Food Science and Technology, University of California, Davis 95616, USA
| | | |
Collapse
|
41
|
Jang YC, Kao LS, Wang FF. Involvement of Ca2+ signalling in the vasoactive intestinal peptide and 8-Br-cAMP induction of c-fos mRNA expression. Cell Signal 1998; 10:27-34. [PMID: 9502114 DOI: 10.1016/s0898-6568(97)00069-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vasoactive intestinal peptide (VIP) is known to signal via Gs mediated pathways. VIP stimulated c-fos mRNA expression in a clonal GH3 pituitary tumour cell line, GH3Ca, whereas 8-Br-cAMP only moderately induced c-fos expression. The VIP-induced c-fos expression was inhibited in the presence of EGTA, or the L-type Ca2+ channel blockers verapamil and nifedipine. Measurement of intracellular Ca2+ concentration ([Ca2+]i) by Fura-2 indicates that VIP gradually elevates [Ca2+]i, with the maximum level attained at 4 min following hormone addition. No [Ca2+]i increase could be detected in Ca2+ free buffer or in buffer containing nifedipine or verapamil, which suggests that VIP induced Ca2+ entry from L-type Ca2+ channels. 8-Br-cAMP rapidly increased [Ca2+]i, with a maximum concentration attained within 1 min of its addition and the elevated level maintained for 15 min. In the absence of external Ca2+ or in the presence of verapamil or nifedipine, the sustained Ca2+ increase was abolished whereas the transient Ca2+ peak was unaffected. Depletion of the internal calcium pools by thapsigargin (1 microM, 30 min), on the other hand, blocked the rapid transient [Ca2+], rise, suggesting the biphasic [Ca2+]i elicited by 8-Br-cAMP was due to mobilization from internal Ca2+ pool followed by extracellular flow. Interestingly, pretreatment with thapsigargin greatly potentiated the 8-Br-cAMP-stimulated c-fos expression. Pretreatment of cells with cholera toxin (1 microg/ml, 9 h) to deplete Gs proteins abolished VIP stimulated-[Ca2+] elevation, while it had little effect on the 8-Br-cAMP induced [Ca2+]i rise. Our results show that VIP increased Ca2+ influx from L-type channel through a Gs-mediated mechanism and this Ca2+ entry across the plasma membrane plays a major role in the hormone induced c-fos mRNA expression.
Collapse
Affiliation(s)
- Y C Jang
- Institute of Biochemistry, National Yang-Ming University, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
42
|
Aebert H, Cornelius T, Ehr T, Holmer SR, Birnbaum DE, Riegger GA, Schunkert H. Expression of immediate early genes after cardioplegic arrest and reperfusion. Ann Thorac Surg 1997; 63:1669-75. [PMID: 9205166 DOI: 10.1016/s0003-4975(97)00272-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Induction of protooncogenes such as c-fos, c-jun, and EGR-1 has been implicated in cellular growth and differentiation. Heat-shock proteins (HSPs) such as hsp 70 may mediate resistance to ischemia after heat shock and ischemic preconditioning. The effects of cardioplegia on the regulation of these immediate early genes are unclear. METHODS Isolated rat hearts were subjected to different cold (5 degrees C) or normothermic (35 degrees C) cardioplegic solutions and reperfused with normothermic Krebs-Henseleit buffer. Right atrial biopsy specimens from patients undergoing coronary artery bypass grafting with cold cardioplegic arrest were taken before and after cardiopulmonary bypass. Analysis of immediate early gene messenger RNAs was performed using Northern blots. Related proteins were localized by immunohistochemistry. RESULTS In rat hearts, cold cardioplegia for 40 minutes with Bretschneider or St. Thomas' II solution followed by 40 minutes' reperfusion resulted in a significant increase in left ventricular c-fos, EGR-1, and c-jun messenger RNA levels (4.0-, 3.1-, and 3.0-fold, respectively, with Bretschneider solution and 3.7-, 2.8-, and 2.1-fold, respectively, with St. Thomas' II solution) compared with control hearts perfused at 35 degrees C with Krebs-Henseleit buffer. Normothermic cardioplegia with St. Thomas' II solution was without effect, whereas sequential perfusion with Krebs-Henseleit buffer at 5 degrees C and 35 degrees C resulted in a similar increase in protooncogene messenger RNA levels. Only cold Bretschneider solution was related to a 5.2-fold induction of hsp 70 messenger RNA levels. Likewise, rat atrial tissues and samples from patients after cardiopulmonary bypass displayed a significant induction of these immediate early genes. Monoclonal antibodies against c-FOS and HSP 70 proteins stained nuclei and perinuclear spaces of endothelial cells and cardiac myocytes. CONCLUSIONS Cold cardioplegic arrest and normothermic reperfusion are potent triggers for immediate early gene induction. Hypothermia emerged as the prime stimulus for the examined protooncogenes. In contrast, hsp 70 induction was dependent on the cardioplegic solution.
Collapse
Affiliation(s)
- H Aebert
- Department of Thoracic and Cardiovascular Surgery, University Hospital, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Funk M, Poensgen B, Graulich W, Jerome V, Müller R. A novel, transformation-relevant activation domain in Fos proteins. Mol Cell Biol 1997; 17:537-44. [PMID: 9001206 PMCID: PMC231778 DOI: 10.1128/mcb.17.2.537] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have previously demonstrated that transformation by Fos is critically dependent on an intact DNA-binding domain (bZip) and a functional N-terminal transactivation motif (N-TM). We now show that a novel motif (C-terminal transactivation motif [C-TM]) near the C terminus also plays an important role in both transformation and the activation of AP1-dependent transcription and that the hydrophobic amino acids in the C-TM are functionally essential. The C-TM is the most crucial element in the C-terminal transactivation domain in Fos, as indicated by its relative strength and context-independent function. The C-TM is clearly different from the previously identified HOB2 domain, located N terminally to the C-TM, and the C-terminally positioned TATA-binding protein-binding domain. We also show that the C-terminal transactivation domain strongly synergizes with the HOB1-like N-TM, even when both domains are present on different proteins within a dimeric complex, and that the C-TM plays a crucial role in this cooperation. These observations can be corroborated in a model in which multiple contacts with the basal machinery are established either to stabilize the transcription complex or to facilitate its sequential assembly.
Collapse
Affiliation(s)
- M Funk
- Institut für Molekularbiologie und Tumorforschung, Phillips-Universität Marburg, Germany
| | | | | | | | | |
Collapse
|
44
|
Aniento F, Papavassiliou AG, Knecht E, Roche E. Selective uptake and degradation of c-Fos and v-Fos by rat liver lysosomes. FEBS Lett 1996; 390:47-52. [PMID: 8706827 DOI: 10.1016/0014-5793(96)00625-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The transcription factor c-Fos is a short-lived protein and calpains and ubiquitin-dependent systems have been proposed to be involved in its degradation. In this report, we consider a lysosomal degradation pathway for c-Fos. Using a cell-free assay, we have found that freshly isolated lysosomes can take up and degrade c-Fos with high efficiency. v-Fos, the oncogenic counterpart of c-Fos, can also be taken up by lysosomes, yet the amount of incorporated protein is much lower. c-Fos uptake is independent of its phosphorylation state but it appears to be regulated by dimerization with differentially phosphorylated forms of c-Jun, while v-Fos escapes this regulation. Moreover, we show that c-Fos is immunologically detected in lysosomes isolated from the liver of rats treated with the protease inhibitor leupeptin. Altogether, these results suggest that lysosomes can also participate in the selective degradation of c-Fos in rat liver.
Collapse
Affiliation(s)
- F Aniento
- Dpt. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Valencia, Burjassot, Spain
| | | | | | | |
Collapse
|
45
|
Chen HH, Liu HM. A new fluorescent histological marker for ischemic neurons, EA 50: correlated with Fos and Jun/AP-1 immunoreactivity. Histochem Cell Biol 1996; 105:375-82. [PMID: 8781991 DOI: 10.1007/bf01463658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cerebral ischemia/hypoxia induces ischemic neuronal changes characterized by nuclear pyknosis, cytoplasmic shrinkage, and basophilia. The ischemic neurons were shown to exhibit strong and persistent c-fos proto-oncogene. The ischemic neuronal changes and c-fos expression are thought to be the consequence of release of excessive glutamate by the ischemic neurons. In the present study, we investigated with immunohisto-chemistry the subcellular distribution of Fos and Jun/AP-1, the protein products of c-fos and c-jun proto-oncogenes, and compared them with histological changes shown by hematoxylin-eosin and by EA 50 stains. The latter is a stain mixture used traditionally in the Papani-colaou procedure and has a specific affinity for ischemic neurons. The active ingredient is eosin Y, a tetrabrominated derivative of fluorescein. With EA 50, the ischemic neurons stain red and emit a yellow fluorescence, while the non-ischemic neurons are green and non-fluorescent. The subcellular site of eosin Y binding corresponds with Fos and Jun/AP-1; all are concentrated in the nuclei and spread into the perikaryon, dendrites, and axons. The eosin Y-binding appears in neurons that have shown advanced ischemic changes. The dye is thus a good histological marker for damaged neurons, but requires freshly fixed tissues and paraffin sections of less than 4 microns thick. Preincubation of tissue sections in antibodies against Fos and Jun abolishes the eosin Y binding, suggesting that the dye may interact with Fos/Jun/AP-1 protein or other protein products in the ischemic neurons.
Collapse
Affiliation(s)
- H H Chen
- Department of Neurosurgery, National Cheng Kung University, Medical College, Tainan, Taiwan, Republic of China
| | | |
Collapse
|
46
|
Ogita K, Amizuka T, Azuma Y, Yoneda Y. Differentiation by magnesium ions of affinities of nuclear proteins for consensus core nucleotide element of the transcription factor c-Myc in murine brain. Neurochem Res 1996; 21:201-9. [PMID: 9182244 DOI: 10.1007/bf02529136] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The addition of divalent cations such as Mg(2+) and Ca(2+) ions markedly reduced binding of a radiolabeled double stranded oligonucleotide probe for the transcription factor c-Myc in the presence of 100 mM KCl in nuclear extracts of the mouse whole brain. Irrespective of the addition of MgCl(2), binding was selectively competed with the unlabeled probe for c-Myc having a double stranded conformation. Treatment with V8 protease differentially modulated binding of the probe for c-Myc determined in the presence and absence of added MgCl(2). Introduction of irreversible covalent bonding between the radiolabeled probe and nuclear proteins led to retarded mobility of the radioactive probe/protein complex in the presence of MgCl(2) on sodium dodecyl sulfate electrophoresis regardless of treatment with DNase. However, an antibody against the c-Myc protein affected neither mobility nor intensity of the radioactive band on gel retardation electrophoresis. Moreover, regional distribution was different from each other in mouse brain when determined in the presence and absence of added MgCl(2). These results suggest that magnesium ions may have an ability to differentiate between nuclear c-Myc family proteins with different affinities for the consensus core nucleotide element CACGTG in murine brain.
Collapse
Affiliation(s)
- K Ogita
- Department of Pharmacology, Setsunan University, Hirakata, Osaka, Japan
| | | | | | | |
Collapse
|
47
|
Stancovski I, Gonen H, Orian A, Schwartz AL, Ciechanover A. Degradation of the proto-oncogene product c-Fos by the ubiquitin proteolytic system in vivo and in vitro: identification and characterization of the conjugating enzymes. Mol Cell Biol 1995; 15:7106-16. [PMID: 8524278 PMCID: PMC230966 DOI: 10.1128/mcb.15.12.7106] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The transcription factor c-Fos is a short-lived cellular protein. The levels of the protein fluctuate significantly and abruptly during changing pathophysiological conditions. Thus, it is clear that degradation of the protein plays an important role in its tightly regulated activity. We examined the involvement of the ubiquitin pathway in c-Fos breakdown. Using a mutant cell line, ts20, that harbors a thermolabile ubiquitin-activating enzyme, E1, we demonstrate that impaired function of the ubiquitin system stabilizes c-Fos in vivo. In vitro, we reconstituted a cell-free system and demonstrated that the protein is multiply ubiquitinated. The adducts serve as essential intermediates for degradation by the 26S proteasome. We show that both conjugation and degradation are significantly stimulated by c-Jun, with which c-Fos forms the active heterodimeric transcriptional activator AP-1. Analysis of the enzymatic cascade involved in the conjugation process reveals that the ubiquitin-carrier protein E2-F1 and its human homolog UbcH5, which target the tumor suppressor p53 for degradation, are also involved in c-Fos recognition. The E2 enzyme acts along with a novel species of ubiquitin-protein ligase, E3. This enzyme is distinct from other known E3s, including E3 alpha/UBR1, E3 beta, and E6-AP. We have purified the novel enzyme approximately 350-fold and demonstrated that it is a homodimer with an apparent molecular mass of approximately 280 kDa. It contains a sulfhydryl group that is essential for its activity, presumably for anchoring activated ubiquitin as an intermediate thioester prior to its transfer to the substrate. Taken together, our in vivo and in vitro studies strongly suggest that c-Fos is degraded in the cell by the ubiquitin-proteasome proteolytic pathway in a process that requires a novel recognition enzyme.
Collapse
Affiliation(s)
- I Stancovski
- Department of Biochemistry, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | | | |
Collapse
|
48
|
Porte D, Oertel-Buchheit P, Granger-Schnarr M, Schnarr M. Fos leucine zipper variants with increased association capacity. J Biol Chem 1995; 270:22721-30. [PMID: 7559397 DOI: 10.1074/jbc.270.39.22721] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The Fos wild-type leucine zipper is unable to support homodimerization. This finding is generally explained by the negative net charge of the Fos zipper leading to the electrostatic repulsion of two monomers. Using a LexA-dependent in vivo assay in Escherichia coli, we show here that additional antideterminants for Fos zipper association are the residues in position a within the Fos zipper interface. If the wild-type Fos zipper is fused to the DNA binding domain of the LexA repressor (LexA-DBD), no excess repression is observed as compared with the LexA-DBD alone, in agreement with the incapacity of the wild-type Fos zipper to promote homodimerization. If hydrophobic amino acids (Ile, Leu, Val, Phe, Met) are inserted into the five a positions of a LexA-Fos zipper fusion protein, substantial transcriptional repression is recovered showing that Fos zipper homodimerization is not only limited by the repulsion of negatively charged residues but also by the nonhydrophobic nature of the a positions. The most efficient variants (harboring Ile or Leu in the five a positions) show an about 80-fold increase in transcriptional repression as compared with the wild-type Fos zipper fusion protein. In the case of multiple identical substitutions, the overall improvement is correlated with the hydrophobicity of the inserted side chains, i.e. Ile Leu > Val > Phe > Met. However at least for Val, Phe, and Met the impact of a given residue type on the association efficiency depends strongly on the heptad, i.e. on the local environment of the a residue. This is particularly striking for the second heptad of the Fos zipper, where Val is less well tolerated than Phe and Met. Most likely the a1 residue modulates the interhelical repulsion between two glutamic acid side chains in positions g1 and e2. Most of the hydrophobic Fos zipper variants are also improved in heteroassociation with a Jun leucine zipper, such that roughly half of the additional free energy of homodimerization is imported into the heterodimer. A few candidates (including the Fos wild-type zipper) deviate from this correlation, showing considerable excess heteroassociation.
Collapse
Affiliation(s)
- D Porte
- Institut de Biologie Moléculaire et Cellulaire du CNRS, UPR 9002, Strasbourg, France
| | | | | | | |
Collapse
|
49
|
van der Burg B, Slager-Davidov R, van der Leede BM, de Laat SW, van der Saag PT. Differential regulation of AP1 activity by retinoic acid in hormone-dependent and -independent breast cancer cells. Mol Cell Endocrinol 1995; 112:143-52. [PMID: 7489817 DOI: 10.1016/0303-7207(95)03600-c] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have studied the role of the AP1 transcription factor in the progression of human breast carcinomas. This progression is characterized by a loss of dependence for proliferation on mitogenic hormones, and is also linked to loss of responsiveness to the growth inhibitor retinoic acid (RA). In the hormone-dependent breast tumor cell line MCF7 mitogenic stimulation was found to be linked to an enhancement of AP1 transcriptional activity, while growth inhibition by RA was parallelled by decreased AP1 activity. AP1 binding activity to its consensus DNA sequence was rapidly reduced in RA treated cells, in the absence of any noticeable change in expression of AP1 constituents. AP1 overexpression abrogated RA repression in MCF7 cells. In hormone-independent cell lines (BT20, Hs578T, MDA-MB231, MDA-MB468) autonomous proliferation was associated with an increased background AP1 activity. Interestingly, these cells are refractory to growth inhibition by RA, which can only be partly explained by underexpression of RA receptors. In these cells RA did not repress AP1 transactivation unless RA receptors were overexpressed by means of cotransfection with an expression vector. This suggests that the high background levels of AP1 activity in the autonomously growing cells are associated with prevention of RA inhibition of AP1 activity to occur. Therefore, increased AP1 activity may not only play a role in progression of breast tumors towards hormone-insensitivity but may also contribute to the RA resistance of such cells.
Collapse
Affiliation(s)
- B van der Burg
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Bassuk AG, Leiden JM. A direct physical association between ETS and AP-1 transcription factors in normal human T cells. Immunity 1995; 3:223-37. [PMID: 7648395 DOI: 10.1016/1074-7613(95)90092-6] [Citation(s) in RCA: 153] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The Ets and AP-1 families of transcription factors bind distinct DNA elements and subserve diverse functions in multiple lymphoid and nonlymphoid cell types. Functionally important Ets and AP-1 binding sites have been identified in a large number of enhancer elements, suggesting important cooperative interactions between these two families of transcription factors. In this report, we have demonstrated a direct physical interaction between Ets and AP-1 proteins both in vitro and in activated human T cells. This interaction is mediated by the binding of the basic domain of Jun to the Ets domain of Ets proteins. Jun, in association with Ets, is capable of interacting with Fos family members to form a trimolecular protein complex. The physical association between Ets-1 and AP-1 proteins is required for the transcriptional activity of enhancer elements containing adjacent Ets and AP-1 binding sites. We conclude that direct physical interactions between Ets and AP-1 transcription factors play an important role in regulating mammalian gene expression.
Collapse
Affiliation(s)
- A G Bassuk
- Department of Medicine, University of Chicago, Illinois 60637, USA
| | | |
Collapse
|