1
|
Sun X, Xiao C, Wang X, Wu S, Yang Z, Sui B, Song Y. Role of post-translational modifications of Sp1 in cancer: state of the art. Front Cell Dev Biol 2024; 12:1412461. [PMID: 39228402 PMCID: PMC11368732 DOI: 10.3389/fcell.2024.1412461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/16/2024] [Indexed: 09/05/2024] Open
Abstract
Specific protein 1 (Sp1) is central to regulating transcription factor activity and cell signaling pathways. Sp1 is highly associated with the poor prognosis of various cancers; it is considered a non-oncogene addiction gene. The function of Sp1 is complex and contributes to regulating extensive transcriptional activity, apart from maintaining basal transcription. Sp1 activity and stability are affected by post-translational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, glycosylation, and SUMOylation. These modifications help to determine genetic programs that alter the Sp1 structure in different cells and increase or decrease its transcriptional activity and DNA binding stability in response to pathophysiological stimuli. Investigating the PTMs of Sp1 will contribute to a deeper understanding of the mechanism underlying the cell signaling pathway regulating Sp1 stability and the regulatory mechanism by which Sp1 affects cancer progression. Furthermore, it will facilitate the development of new drug targets and biomarkers, thereby elucidating considerable implications in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Chinese Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyang Wang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Wu
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhendong Yang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bowen Sui
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
2
|
Focus on Cdc42 in Breast Cancer: New Insights, Target Therapy Development and Non-Coding RNAs. Cells 2019; 8:cells8020146. [PMID: 30754684 PMCID: PMC6406589 DOI: 10.3390/cells8020146] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Breast cancer is the most common malignant tumors in females. Although the conventional treatment has demonstrated a certain effect, some limitations still exist. The Rho guanosine triphosphatase (GTPase) Cdc42 (Cell division control protein 42 homolog) is often upregulated by some cell surface receptors and oncogenes in breast cancer. Cdc42 switches from inactive guanosine diphosphate (GDP)-bound to active GTP-bound though guanine-nucleotide-exchange factors (GEFs), results in activation of signaling cascades that regulate various cellular processes such as cytoskeletal changes, proliferation and polarity establishment. Targeting Cdc42 also provides a strategy for precise breast cancer therapy. In addition, Cdc42 is a potential target for several types of non-coding RNAs including microRNAs and lncRNAs. These non-coding RNAs is extensively involved in Cdc42-induced tumor processes, while many of them are aberrantly expressed. Here, we focus on the role of Cdc42 in cell morphogenesis, proliferation, motility, angiogenesis and survival, introduce the Cdc42-targeted non-coding RNAs, as well as present current development of effective Cdc42-targeted inhibitors in breast cancer.
Collapse
|
3
|
Scheraga RG, Thompson C, Tulapurkar ME, Nagarsekar AC, Cowan M, Potla R, Sun J, Cai R, Logun C, Shelhamer J, Todd NW, Singh IS, Luzina IG, Atamas SP, Hasday JD. Activation of heat shock response augments fibroblast growth factor-1 expression in wounded lung epithelium. Am J Physiol Lung Cell Mol Physiol 2016; 311:L941-L955. [PMID: 27638903 DOI: 10.1152/ajplung.00262.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
We previously showed that coincident exposure to heat shock (HS; 42°C for 2 h) and TNF-α synergistically induces apoptosis in mouse lung epithelium. We extended this work by analyzing HS effects on human lung epithelial responses to clinically relevant injury. Cotreatment with TNF-α and HS induced little caspase-3 and poly(ADP-ribose) polymerase cleavage in human small airway epithelial cells, A549 cells, and BEAS2B cells. Scratch wound closure rates almost doubled when A549 and BEAS2B cells and air-liquid interface cultures of human bronchial epithelial cells were heat shocked immediately after wounding. Microarray, qRT-PCR, and immunoblotting showed fibroblast growth factor 1 (FGF1) to be synergistically induced by HS and wounding. Enhanced FGF1 expression in HS/wounded A549 was blocked by inhibitors of p38 MAPK (SB203580) or HS factor (HSF)-1 (KNK-437) and in HSF1 knockout BEAS2B cells. PCR demonstrated FGF1 to be expressed from the two most distal promoters in wounded/HS cells. Wound closure in HS A549 and BEAS2B cells was reduced by FGF receptor-1/3 inhibition (SU-5402) or FGF1 depletion. Exogenous FGF1 accelerated A549 wound closure in the absence but not presence of HS. In the presence of exogenous FGF1, HS slowed wound closure, suggesting that it increases FGF1 expression but impairs FGF1-stimulated wound closure. Frozen sections from normal and idiopathic pulmonary fibrosis (IPF) lung were analyzed for FGF1 and HSP70 by immunofluorescence confocal microscopy and qRT-PCR. FGF1 and HSP70 mRNA levels were 7.5- and 5.9-fold higher in IPF than normal lung, and the proteins colocalized to fibroblastic foci in IPF lung. We conclude that HS signaling may have an important impact on gene expression contributing to lung injury, healing, and fibrosis.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | | | - Mohan E Tulapurkar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashish C Nagarsekar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark Cowan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ratnakar Potla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Junfeng Sun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Rongman Cai
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Carolea Logun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - James Shelhamer
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Nevins W Todd
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ishwar S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Irina G Luzina
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Sergei P Atamas
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; .,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| |
Collapse
|
4
|
Marwa BAG, Raguema N, Zitouni H, Feten HBA, Olfa K, Elfeleh R, Almawi W, Mahjoub T. FGF1 and FGF2 mutations in preeclampsia and related features. Placenta 2016; 43:81-5. [PMID: 27324104 DOI: 10.1016/j.placenta.2016.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/08/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND Fibroblast growth factor (FGF) 1 and FGF2 were previously linked with preeclampsia (PE), possibly through altering decidual and placental FGFR2 expression. Since common variation in FGF1 and FGF2 might influence FGF1 and FGF2 activity, this study evaluated whether common FGF1 and FGF2 variants are linked with PE and associated features. METHODS The association between FGF1 rs34011 and FGF2 rs2922979 SNPs and PE were tested in 300 women with PE, and 300 age-matched control women. RESULTS The allelic distribution of FGF1 rs34011 (P < 0.001) but not FGF2 rs2922979, variants were significantly different between PE cases and control women. Marginal association of FGF2 rs2922979 was seen after controlling for key covariates. Setting homozygous major allele genotype (1/1) as reference, significantly higher frequencies of heterozygous rs345011, and reduced frequency of heterozygous rs2922979 genotype carriers were seen in PE cases; the distribution of the remaining genotypes were comparable between cases and controls. Carriage of rs2922979 minor allele correlated with fasting glucose (P = 0.02), while the presence of rs34011 minor allele was not correlated with PE-associated features. CONCLUSIONS Our study suggests that the genetic variants of FGF1 rs34011, more so than FGF2 rs2922979, may play a role in PE pathogenesis in Tunisian women. These findings need confirmation in other ethnic populations.
Collapse
Affiliation(s)
- Ben Ali Gannoun Marwa
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Tunisia; Faculty of Science of Bizerte, University of Carthage, Tunisia.
| | - Nozha Raguema
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Tunisia; Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Hedia Zitouni
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Tunisia; Faculty of Science of Bizerte, University of Carthage, Tunisia
| | - Hachani Ben Ali Feten
- University Hospital F. Hached, Department of Obstetrics and Gynaecology, Sousse, Tunisia
| | - Kacem Olfa
- University Hospital F. Hached, Department of Obstetrics and Gynaecology, Sousse, Tunisia
| | - Raja Elfeleh
- Centre of Maternity and Neonatology, Monastir, Tunisia
| | - Wassim Almawi
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 22979, Manama, Bahrain
| | - Touhami Mahjoub
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy of Monastir, University of Monastir, Tunisia
| |
Collapse
|
5
|
FGF1 C-terminal domain and phosphorylation regulate intracrine FGF1 signaling for its neurotrophic and anti-apoptotic activities. Cell Death Dis 2016; 7:e2079. [PMID: 26844696 PMCID: PMC4849156 DOI: 10.1038/cddis.2016.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 12/17/2015] [Accepted: 12/23/2015] [Indexed: 11/09/2022]
Abstract
Fibroblast growth factor 1 (FGF1) is a prototypic member of the FGFs family overexpressed in various tumors. Contrarily to most FGFs, FGF1 lacks a secretion peptide signal and acts mainly in an intracellular and nuclear manner. Intracellular FGF1 induces cell proliferation, differentiation and survival. We previously showed that intracellular FGF1 induces neuronal differentiation and inhibits both p53- and serum-free-medium-induced apoptosis in PC12 cells. FGF1 nuclear localization is required for these intracellular activities, suggesting that FGF1 regulates p53-dependent apoptosis and neuronal differentiation by new nuclear pathways. To better characterize intracellular FGF1 pathways, we studied the effect of three mutations localized in the C-terminal domain of FGF1 (i.e., FGF1K132E, FGF1S130A and FGF1S130D) on FGF1 neurotrophic and anti-apoptotic activities in PC12 cells. The change of the serine 130 to alanine precludes FGF1 phosphorylation, while its mutation to aspartic acid mimics phosphorylation. These FGF1 mutants kept both a nuclear and cytosolic localization in PC12 cells. Our study highlights for the first time the role of FGF1 phosphorylation and the implication of FGF1 C-terminal domain on its intracellular activities. Indeed, we show that the K132E mutation inhibits both the neurotrophic and anti-apoptotic activities of FGF1, suggesting a regulatory activity for FGF1 C terminus. Furthermore, we observed that both FGF1S130A and FGF1S130D mutant forms induced PC12 cells neuronal differentiation. Therefore, FGF1 phosphorylation does not regulate FGF1-induced differentiation of PC12 cells. Then, we showed that only FGF1S130A protects PC12 cells against p53-dependent apoptosis, thus phosphorylation appears to inhibit FGF1 anti-apoptotic activity in PC12 cells. Altogether, our results show that phosphorylation does not regulate FGF1 neurotrophic activity but inhibits its anti-apoptotic activity after p53-dependent apoptosis induction, giving new insight into the poorly described FGF1 intracrine/nuclear pathway. The study of nuclear pathways could be crucial to identify key regulators involved in neuronal differentiation, tumor progression and resistances to radio- and chemo-therapy.
Collapse
|
6
|
Promoter-Dependent Translation Controlled by p54nrb and hnRNPM during Myoblast Differentiation. PLoS One 2015; 10:e0136466. [PMID: 26332123 PMCID: PMC4558007 DOI: 10.1371/journal.pone.0136466] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/04/2015] [Indexed: 01/08/2023] Open
Abstract
Fibroblast growth factor 1 (FGF1) is induced during myoblast differentiation at both transcriptional and translational levels. Here, we identify hnRNPM and p54nrb/NONO present in protein complexes bound to the FGF1 promoter and to the mRNA internal ribosome entry site (IRES). Knockdown or overexpression of these proteins indicate that they cooperate in activating IRES-dependent translation during myoblast differentiation, in a promoter-dependent manner. Importantly, mRNA transfection and promoter deletion experiments clearly demonstrate the impact of the FGF1 promoter on the activation of IRES-dependent translation via p54nrb and hnRNPM. Accordingly, knockdown of either p54 or hnRNPM also blocks endogenous FGF1 induction and myotube formation, demonstrating the physiological relevance of this mechanism and the role of these two proteins in myogenesis. Our study demonstrates the cooperative function of hnRNPM and p54nrb as regulators of IRES-dependent translation and indicates the involvement of a promoter-dependent mechanism.
Collapse
|
7
|
Beishline K, Azizkhan-Clifford J. Sp1 and the 'hallmarks of cancer'. FEBS J 2015; 282:224-58. [PMID: 25393971 DOI: 10.1111/febs.13148] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 09/26/2014] [Accepted: 11/10/2014] [Indexed: 12/19/2022]
Abstract
For many years, transcription factor Sp1 was viewed as a basal transcription factor and relegated to a role in the regulation of so-called housekeeping genes. Identification of Sp1's role in recruiting the general transcription machinery in the absence of a TATA box increased its importance in gene regulation, particularly in light of recent estimates that the majority of mammalian genes lack a TATA box. In this review, we briefly consider the history of Sp1, the founding member of the Sp family of transcription factors. We review the evidence suggesting that Sp1 is highly regulated by post-translational modifications that positively and negatively affect the activity of Sp1 on a wide array of genes. Sp1 is over-expressed in many cancers and is associated with poor prognosis. Targeting Sp1 in cancer treatment has been suggested; however, our review of the literature on the role of Sp1 in the regulation of genes that contribute to the 'hallmarks of cancer' illustrates the extreme complexity of Sp1 functions. Sp1 both activates and suppresses the expression of a number of essential oncogenes and tumor suppressors, as well as genes involved in essential cellular functions, including proliferation, differentiation, the DNA damage response, apoptosis, senescence and angiogenesis. Sp1 is also implicated in inflammation and genomic instability, as well as epigenetic silencing. Given the apparently opposing effects of Sp1, a more complete understanding of the function of Sp1 in cancer is required to validate its potential as a therapeutic target.
Collapse
Affiliation(s)
- Kate Beishline
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
8
|
Kang S, Li SZ, Wang N, Zhou RM, Wang T, Wang DJ, Li XF, Bui J, Li Y. Association between genetic polymorphisms in fibroblast growth factor (FGF)1 and FGF2 and risk of endometriosis and adenomyosis in Chinese women. Hum Reprod 2010; 25:1806-11. [PMID: 20504870 DOI: 10.1093/humrep/deq128] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Angiogenesis appears to be an important event in the pathophysiology of endometriosis (EM) and adenomyosis. Two angiogenic factors, fibroblast growth factor (FGF) 1 and 2, play a central role in the initiation of angiogenesis. We investigated whether FGF1 -1385A/G and FGF2 754C/G polymorphisms are associated with a risk of developing EM and adenomyosis. METHODS Genotypes were analyzed by the PCR-restriction fragment length polymorphism method in two groups of women, of Han ethnicity in north China, aged 16-55 years: (1) 421 EM patients and 421 controls; (2) 269 adenomyosis patients and 269 controls. RESULTS There was no difference in genotype distribution of the FGF1 -1385A/G polymorphism between adenomyosis cases and controls (P > 0.05), but the frequency of the A allele in EM patients was lower than that in controls (P = 0.013). Genotype and allele frequencies of the FGF2 754C/C polymorphism were significantly different in both EM and adenomyosis cases versus control groups. Compared with C/C homozygotes, the G allele (C/G + G/G) was associated with a decreased susceptibility to developing EM [odds ratio (OR) = 0.575, 95% confidence interval (CI) = 0.387-0.854] and adenomyosis (OR = 0.577, 95% CI = 0.367-0.906). Combined genotype analysis of both polymorphisms also showed differences between cases versus controls (all P < 0.001). CONCLUSIONS Our study shows for the first time that the FGF2 754C/G polymorphism may be associated with a risk of developing EM and adenomyosis in north Chinese women. Carriers of the G allele in the FGF2 gene appear to be protected from these gynecological diseases. Further studies in other populations, and of other candidate genes, are now warranted.
Collapse
Affiliation(s)
- Shan Kang
- Department of Obstetrics and Gynaecology, Hebei Cancer Institute, Hebei Medical University, Fourth Hospital, Jiankanglu 12, Shijiazhuang 050011, China
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Bian JT, Zhao HL, Zhang ZX, Bi XH, Zhang JW. No association of the C>T polymorphism that is located 1385 upstream from initial code of fibroblast growth factor 1 gene with Alzheimer's disease in Chinese. Brain Res 2010; 1328:113-7. [DOI: 10.1016/j.brainres.2010.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 03/08/2010] [Accepted: 03/09/2010] [Indexed: 10/19/2022]
|
10
|
|
11
|
Abstract
The Rho family of GTPases is part of the Ras superfamily. The Rho, Rac, and Cdc42 members of the family are present in mammalian cells and have been the subject of attention of researchers due to their vast spectrum of functions. Rac 1, Cdc42, and RhoA are well-known for their role in the regulation of the actin cytoskeleton in promoting the formation of lamellipodia, filopodia, and stress fibers, respectively. The Rho proteins also participate in the control of cell growth, motility, cell-cell adhesions, morphogenesis, cytoskeletal dynamics, and cellular trafficking. The mechanisms for eliciting these functions have become clearer during the last decade. Concordant with their roles in multiple processes of cellular control, the Rho proteins have been shown to be involved in tumor growth, progression, metastasis, and now angiogenesis.
Collapse
Affiliation(s)
- Sofia D Merajver
- Breast and Ovarian Cancer Risk Evaluation Program, University of Michigan Comprehensive Cancer Center, 7217 CCGC, 1500 East Medical Center Drive, Ann Arbor, MI 48109-0948, USA.
| | | |
Collapse
|
12
|
Su ZZ, Sarkar D, Emdad L, Duigou GJ, Young CSH, Ware J, Randolph A, Valerie K, Fisher PB. Targeting gene expression selectively in cancer cells by using the progression-elevated gene-3 promoter. Proc Natl Acad Sci U S A 2005; 102:1059-64. [PMID: 15647352 PMCID: PMC545837 DOI: 10.1073/pnas.0409141102] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2004] [Indexed: 02/07/2023] Open
Abstract
One impediment to effective cancer-specific gene therapy is the rarity of regulatory sequences targeting gene expression selectively in tumor cells. Although many tissue-specific promoters are recognized, few cancer-selective gene promoters are available. Progression-elevated gene-3 (PEG-3) is a rodent gene identified by subtraction hybridization that displays elevated expression as a function of transformation by diversely acting oncogenes, DNA damage, and cancer cell progression. The promoter of PEG-3, PEG-Prom, displays robust expression in a broad spectrum of human cancer cell lines with marginal expression in normal cellular counterparts. Whereas GFP expression, when under the control of a CMV promoter, is detected in both normal and cancer cells, when GFP is expressed under the control of the PEG-Prom, cancer-selective expression is evident. Mutational analysis identifies the AP-1 and PEA-3 transcription factors as primary mediators of selective, cancer-specific expression of the PEG-Prom. Synthesis of apoptosis-inducing genes, under the control of the CMV promoter, inhibits the growth of both normal and cancer cells, whereas PEG-Prom-mediated expression of these genes kills only cancer cells and spares normal cells. The efficacy of the PEG-Prom as part of a cancer gene therapeutic regimen is further documented by in vivo experiments in which PEG-Prom-controlled expression of an apoptosis-inducing gene completely inhibited prostate cancer xenograft growth in nude mice. These compelling observations indicate that the PEG-Prom, with its cancer-specific expression, provides a means of selectively delivering genes to cancer cells, thereby providing a crucial component in developing effective cancer gene therapies.
Collapse
Affiliation(s)
- Zhao-Zhong Su
- Department of Pathology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Wang J, Stockton DW, Ittmann M. The fibroblast growth factor receptor-4 Arg388 allele is associated with prostate cancer initiation and progression. Clin Cancer Res 2005; 10:6169-78. [PMID: 15448004 DOI: 10.1158/1078-0432.ccr-04-0408] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Increased expression of fibroblast growth factors that can activate the fibroblast growth factor receptor-4 (FGFR-4) occurs in a substantial fraction of human prostate cancers in vivo. A germline polymorphism of the FGFR-4 gene resulting in expression of arginine at codon 388 (Arg388) is associated with aggressive disease in patients with breast and colon cancer. We therefore sought to determine whether the FGFR-4 Arg388 allele was associated with prostate cancer incidence and/or the occurrence of aggressive disease. EXPERIMENTAL DESIGN The FGFR-4 genotype of men undergoing radical prostatectomy and controls of the same race was determined and the genotype correlated with clinical and pathologic markers of disease aggressiveness. PNT1A cell lines expressing predominantly the FGFR-4 Arg388 or Gly388 allele were established, and cell migration and invasiveness of these cells were assessed by a wounding assay and by quantitative determination of invasion through Matrigel. Expression of urokinase-type plasminogen activator receptor was determined by quantitative RT-PCR and enzyme-linked immunoabsorption assay. RESULTS Homozygosity for the FGFR-4 Arg388 allele is strongly associated with the occurrence of prostate cancer in white men. The presence of the FGFR-4 Arg388 allele is also correlated with the occurrence of pelvic lymph node metastasis and biochemical (prostate-specific antigen) recurrence. Expression of FGFR-4 Arg388 in immortalized prostatic epithelial cells results in increased cell motility and invasion through Matrigel and was associated with increased expression of urokinase-type plasminogen activator receptor. CONCLUSION The FGFR-4 Arg388 allele is associated with both an increased incidence and clinical aggressiveness of prostate cancer and results in changes in cellular motility and invasiveness in immortalized prostate epithelial cells consistent with the promotion of metastasis.
Collapse
Affiliation(s)
- Jianghua Wang
- Department of Pathology, Baylor College of Medicine, USA
| | | | | |
Collapse
|
14
|
Yamagata H, Chen Y, Akatsu H, Kamino K, Ito JI, Yokoyama S, Yamamoto T, Kosaka K, Miki T, Kondo I. Promoter polymorphism in fibroblast growth factor 1 gene increases risk of definite Alzheimer's disease. Biochem Biophys Res Commun 2004; 321:320-3. [PMID: 15358178 DOI: 10.1016/j.bbrc.2004.06.142] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2004] [Indexed: 11/22/2022]
Abstract
Fibroblast growth factor 1 (FGF1, also known as acidic FGF) protects selective neuronal populations against neurotoxic effects such as those in Alzheimer's disease (AD) and HIV encephalitis. The FGF1 gene is therefore a strong candidate gene for AD. Using the promoter polymorphism of the FGF1 gene, we examined the relationship between AD and the FGF1 and apolipoprotein E (APOE) genes in 100 Japanese autopsy-confirmed late-onset AD patients and 106 age-matched non-demented controls. The promoter polymorphism (-1385 A/G) was significantly associated with AD risk. The odds ratio for AD associated with the GG vs non-GG genotype was 2.02 (95% CI = 1.16-3.52), while that of s4 vs non-ł4 in APOE4 gene was 5.19 (95% CI = 2.68-10.1). The odds ratio for APOEP4 and FGF1 GG carriers was 20.5 (95% CI = 6.88-60.9). The results showed that the FGF1 gene is associated with autopsy-confirmed AD.
Collapse
Affiliation(s)
- Hidehisa Yamagata
- Department of Medical Genetics, Ehime University School of Medicine, Ehime, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Martineau Y, Le Bec C, Monbrun L, Allo V, Chiu IM, Danos O, Moine H, Prats H, Prats AC. Internal ribosome entry site structural motifs conserved among mammalian fibroblast growth factor 1 alternatively spliced mRNAs. Mol Cell Biol 2004; 24:7622-35. [PMID: 15314170 PMCID: PMC507008 DOI: 10.1128/mcb.24.17.7622-7635.2004] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fibroblast growth factor 1 (FGF-1) is a powerful angiogenic factor whose gene structure contains four promoters, giving rise to a process of alternative splicing resulting in four mRNAs with alternative 5' untranslated regions (5' UTRs). Here we have identified, by using double luciferase bicistronic vectors, the presence of internal ribosome entry sites (IRESs) in the human FGF-1 5' UTRs, particularly in leaders A and C, with distinct activities in mammalian cells. DNA electrotransfer in mouse muscle revealed that the IRES present in the FGF-1 leader A has a high activity in vivo. We have developed a new regulatable TET OFF bicistronic system, which allowed us to rule out the possibility of any cryptic promoter in the FGF-1 leaders. FGF-1 IRESs A and C, which were mapped in fragments of 118 and 103 nucleotides, respectively, are flexible in regard to the position of the initiation codon, making them interesting from a biotechnological point of view. Furthermore, we show that FGF-1 IRESs A of murine and human origins show similar IRES activity profiles. Enzymatic and chemical probing of the FGF-1 IRES A RNA revealed a structural domain conserved among mammals at both the nucleotide sequence and RNA structure levels. The functional role of this structural motif has been demonstrated by point mutagenesis, including compensatory mutations. These data favor an important role of IRESs in the control of FGF-1 expression and provide a new IRES structural motif that could help IRES prediction in 5' UTR databases.
Collapse
Affiliation(s)
- Yvan Martineau
- Institut National de la Santé et de la Recherche Médicale U589, Hormones, Facteurs de Croissance et Physiopathologie Vasculaire, Institut Louis Bugnard, IFR31, CHU Rangueil, 31059 Toulouse Cedex 09, France
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kwabi-Addo B, Wang J, Erdem H, Vaid A, Castro P, Ayala G, Ittmann M. The expression of Sprouty1, an inhibitor of fibroblast growth factor signal transduction, is decreased in human prostate cancer. Cancer Res 2004; 64:4728-35. [PMID: 15256439 DOI: 10.1158/0008-5472.can-03-3759] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A considerable body of evidence indicates that alterations of fibroblast growth factors (FGFs) and their receptors contribute to prostate cancer progression. Recently, a new family of regulators of FGF activity has been identified. The Sprouty gene family negatively regulates FGF signaling in a variety of systems and could potentially limit the biological activity of FGFs in prostate cancer. Immunohistochemical analysis of normal and neoplastic prostate tissues using tissue microarrays revealed that Sprouty1 protein is down-regulated in approximately 40% of prostate cancers when compared with matched normal prostate. By quantitative real-time PCR analysis, we found that Sprouty1 mRNA levels were significantly decreased in prostate cancers in vivo in comparison with normal prostate. In prostate cancer cell lines, there is loss of the normal up-regulation of Sprouty1 mRNA and protein in response to FGFs. The decrease in Sprouty1 expression in the human prostate cancer, despite elevated levels of FGF ligands and FGF receptors, implies a loss of an important growth regulatory mechanism in prostate cancers that may potentiate the effects of increased FGF and FGF receptor expression in prostate cancer.
Collapse
|
17
|
Aoki D, Yamamoto-Fukuda T, Hishikawa Y, Nakamura M, Sakai H, Kanetake H, Koji T. Coexpression of Keratinocyte Growth Factor and Its Receptor in Normal and Prostate Cancer Tissues: Possible Formation of Autonomous Andromedin Loop. Acta Histochem Cytochem 2004. [DOI: 10.1267/ahc.37.379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Daiyu Aoki
- Division of Histology and Cell Biology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences
- Division of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences
| | - Tomomi Yamamoto-Fukuda
- Otorhinolaryngology, Department of Translational Medical Science, Nagasaki University Graduate School of Biomedical Sciences
| | - Yoshitaka Hishikawa
- Division of Histology and Cell Biology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences
| | - Mitsuru Nakamura
- Biosciences Research and Development Center (M.N), Nichirei Corporation
| | - Hideki Sakai
- Division of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences
| | - Hiroshi Kanetake
- Division of Nephro-Urology, Nagasaki University Graduate School of Biomedical Sciences
| | - Takehiko Koji
- Division of Histology and Cell Biology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences
| |
Collapse
|
18
|
Hague S, Manek S, Oehler MK, MacKenzie IZ, Bicknell R, Rees MCP. Tamoxifen induction of angiogenic factor expression in endometrium. Br J Cancer 2002; 86:761-7. [PMID: 11875740 PMCID: PMC2375303 DOI: 10.1038/sj.bjc.6600157] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2001] [Accepted: 12/14/2001] [Indexed: 11/09/2022] Open
Abstract
Tamoxifen is the current therapy of choice for patients with oestrogen receptor positive breast cancer, and it is currently under evaluation as a prophylactic for women at high risk of developing the disease. However, tamoxifen is also known to induce proliferative changes in the endometrium increasing the risk of developing endometrial hyperplasia, polyps and carcinoma. Angiogenesis is an intimate part of this process. For this reason, we have examined the expression of several well characterized angiogenic factors, namely, acidic and basic fibroblast growth factor, thymidine phosphorylase, vascular endothelial growth factor and adrenomedullin in both normal and tamoxifen exposed pre- and postmenopausal endometrium. Vascular density and endothelial proliferation index were also quantified. We found increased expression of acidic and basic fibroblast growth factor and adrenomedullin after treatment with tamoxifen mainly in premenopausal tissue. Vascular density was significantly increased in pre- but not post-menopausal endometrium (P=0.0018) following tamoxifen treatment. These results support the notion that angiogenesis is integral to the response to tamoxifen exposure, and is a potential target with which to block these side effects of tamoxifen.
Collapse
Affiliation(s)
- S Hague
- Nuffield Department of Obstetrics and Gynaecology, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | | | | | |
Collapse
|
19
|
Chiu IM, Touhalisky K, Baran C. Multiple controlling mechanisms of FGF1 gene expression through multiple tissue-specific promoters. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2002; 70:155-74. [PMID: 11642361 DOI: 10.1016/s0079-6603(01)70016-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
We now know that fibroblast growth factor-1 (FGF1) transcription is controlled by at least four distinct promoters in a tissue-specific manner. Thus, promoter 1.A is active in the kidney, 1.B in the brain, and 1.C and 1.D in a variety of cultured cells including vascular smooth muscle cells. These promoters are separated from each other by up to 70 kbp. Multiple FGF1 transcripts arise from alternate promoter usage and alternative splicing of different 5'-untranslated exons. The 1.A and 1.B promoters are constitutively active in their respective cell types. In contrast, different biological response modifiers, including serum and transforming growth factor beta, can induce the 1.C and 1.D promoters. The 540-bp sequence upstream of the 1B transcription initiation site is sufficient to drive the expression of a heterologous luciferase reporter in cultured cells, and an 18-bp sequence within this region is important for the regulation of brain-specific gene expression. Furthermore, regulation occurs through the binding of the 18-bp sequence to a brain-specific 37-kDa protein and a ubiquitous basic helix-loop-helix protein, E2-2. We have produced transgenic mice bearing the brain-specific promoter of the human FGF1 gene joined to the SV40 immediate-early gene, which encodes the large T antigen. The resulting mice developed brain tumors that originated in the pontine gray, just rostral to the fourth ventricle. We have also identified a serum response element, comprising a CarG box and an Ets-binding site, in the 1.D promoter. Continued characterization of the mechanistic events that control the tissue-specific activation of FGF1 promoters will help us to understand the role of FGF1 in cancer, atherosclerosis, and neural development.
Collapse
Affiliation(s)
- I M Chiu
- Department of Internal Medicine, The Ohio State University, Columbus 43210, USA
| | | | | |
Collapse
|
20
|
Ozen M, Giri D, Ropiquet F, Mansukhani A, Ittmann M. Role of fibroblast growth factor receptor signaling in prostate cancer cell survival. J Natl Cancer Inst 2001; 93:1783-90. [PMID: 11734594 DOI: 10.1093/jnci/93.23.1783] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Expression of fibroblast growth factors (FGFs) is increased in a substantial fraction of human prostate cancers in vivo and in prostate cancer cell lines. Altered FGF signaling can potentially have a variety of effects, including stimulating cell proliferation and inhibiting cell death. To determine the biologic significance of altered FGF signaling in human prostate cancer, we disrupted signaling by expression of a dominant-negative (DN) FGF receptor in prostate cancer cell lines. METHODS PC-3, LNCaP, and DU145 prostate cancer cells were stably transfected with DN FGFR constructs, and LNCaP and DU145 cells were infected with a recombinant adenovirus expressing DN FGFR-1. The effect of DN FGFR-1 expression was assessed by colony-formation assays, cell proliferation assays, flow cytometry, and cytogenetic analysis. Key regulators involved in the G(2)-to-M cell cycle transition were assessed by western blotting to examine cyclin B1 expression and by in vitro kinase assay to assess cdc2 kinase activity. RESULTS Stable transfection of the DN FGFR-1 construct inhibited colony formation by more than 99% in all three cell lines. Infection of LNCaP and DU145 prostate cancer cells with adenovirus expressing DN FGFR-1 led to extensive cell death within 48 hours. Flow cytometry and cytogenetic analysis revealed that the DN FGFR-1 receptor led to arrest in the G(2) phase of the cell cycle before cell death. Cyclin B1 accumulated in DN FGFR-1-infected LNCaP cells, but cdc2 kinase activity was decreased. CONCLUSIONS These findings reveal an unexpected dependence of prostate cancer cells on FGF receptor signal transduction to traverse the G(2)/M checkpoint. The mechanism for the G(2) arrest is not clear. Our results raise the possibility that FGF-signaling antagonists might enhance the cell death induced by other prostate cancer therapies.
Collapse
Affiliation(s)
- M Ozen
- Department of Pathology, Baylor College of Medicine, 2002 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
21
|
Chotani MA, Touhalisky K, Chiu IM. The small GTPases Ras, Rac, and Cdc42 transcriptionally regulate expression of human fibroblast growth factor 1. J Biol Chem 2000; 275:30432-8. [PMID: 10849427 DOI: 10.1074/jbc.m003545200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Four distinct promoters (1A, 1B, 1C, and 1D) of fibroblast growth factor 1 (FGF1), spaced up to 70 kilobase pairs apart, direct the expression of alternatively spliced transcript variants (FGF1.A, -1. B, -1.C, and -1.D) that encode FGF1. These FGF1 transcripts can be detected in cultured cells as well as in normal and diseased tissues. These transcripts are differentially regulated in a cell-specific manner. To further delineate the biological function of multiple promoter usage by a single gene, we investigated the transcriptional regulation of these promoters by defined signaling pathways associated with cell proliferation and cell survival. Here we show a specific association of two of the FGF1 promoters, 1C and 1D, with signaling cascades of the Ras superfamily of GTPases. A serum-response element, comprised of the Ets and CArG motifs, present in promoter 1D was shown to be the target of distinct signaling cascades; the Ets motif target of Ras, Rac1, and Cdc42 regulation; and the CArG motif target of de novo protein synthesis-independent cascade. Ras and Rac1 also activated the FGF2 promoter. Further, the transcription factor Ets2 synergistically activated FGF1 gene, but not FGF2, in a Ras- and Rac1-dependent signaling pathway. In support of these conclusions high levels of intracellular FGF1 were detected in cells undergoing cytokinesis. Altogether, our results suggest that FGF1 may play a fundamental role in cell division, spreading, and migration, in addition to cell proliferation.
Collapse
Affiliation(s)
- M A Chotani
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio 43210-1228, USA
| | | | | |
Collapse
|