1
|
Chen Y, Jing L, Meng Q, Li B, Chen R, Sun Z. Supramolecular Chemotherapy: Noncovalent Bond Synergy of Cucurbit[7]uril against Human Colorectal Tumor Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:9547-9552. [PMID: 34333979 DOI: 10.1021/acs.langmuir.1c01422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Supramolecular chemotherapy has drawn increasing interest due to its ability to improve the efficiency of antitumor drugs and fewer associated toxic side effects. In this study, the smart supramolecular cargo, the doxorubicin-ZnO-cucurbit[7]uril (CDZ) nanocomplex, was constructed through ion-dipole interactions between cucurbit[7]uril {CB[7]} and doxorubicin-ZnO (dox-ZnO). The binding affinity of CB[7] and dox-ZnO was determined to be 104 M-1 by isothermal titration calorimetry. Importantly, spermine had a stronger binding affinity (106 M-1) with CB[7] than dox-ZnO through host-guest interactions. In the tumor microenvironment, spermine disassembled the CDZ nanocomplex, and dox was released from the nanocomplex by XRD, UV-visible spectra, and contact angle analysis. Compared to the single drug dox, the CDZ nanocomplex was demonstrated to possess higher activity of killing colorectal tumor cells by confocal laser scanning microscopy and cytotoxicity, which could be attributed to spermine concentration, spermine synthase, free radical damage, and G1 cell cycle arrest. Overall, the supramolecular delivery of dox can enhance the inhibition of human colorectal tumor cell proliferation and reduce cytotoxicity in human myocardial cells through the noncovalent bond synergy of {CB[7]}.
Collapse
Affiliation(s)
- Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Li Jing
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Qingtao Meng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Bin Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Rui Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environment Toxicology, Capital Medical University, Beijing 100069, P. R. China
| |
Collapse
|
2
|
Gamage TKJB, Schierding W, Hurley D, Tsai P, Ludgate JL, Bhoothpur C, Chamley LW, Weeks RJ, Macaulay EC, James JL. The role of DNA methylation in human trophoblast differentiation. Epigenetics 2018; 13:1154-1173. [PMID: 30475094 DOI: 10.1080/15592294.2018.1549462] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The placenta is a vital fetal exchange organ connecting mother and baby. Specialised placental epithelial cells, called trophoblasts, are essential for adequate placental function. Trophoblasts transform the maternal vasculature to allow efficient blood flow to the placenta and facilitate adequate nutrient uptake. Placental development is in part regulated by epigenetic mechanisms. However, our understanding of how DNA methylation contributes to human trophoblast differentiation is limited. To better understand how genome-wide methylation differences affect trophoblast differentiation, reduced representation bisulfite sequencing (RRBS) was conducted on four matched sets of trophoblasts; side-population trophoblasts (a candidate human trophoblast stem cell population), cytotrophoblasts (an intermediate progenitor population), and extravillous trophoblasts (EVT, a terminally differentiated population) each isolated from the same first trimester placenta. Each trophoblast population had a distinct methylome. In line with their close differentiation relationship, the methylation profile of side-population trophoblasts was most similar to cytotrophoblasts, whilst EVT had the most distinct methylome. In comparison to mature trophoblast populations, side-population trophoblasts exhibited differential methylation of genes and miRNAs involved in cell cycle regulation, differentiation, and regulation of pluripotency. A combined methylomic and transcriptomic approach was taken to better understand cytotrophoblast differentiation to EVT. This revealed methylation of 41 genes involved in epithelial to mesenchymal transition and metastatic cancer pathways, which likely contributes to the acquisition of an invasive EVT phenotype. However, the methylation status of a gene did not always predict gene expression. Therefore, while CpG methylation plays a role in trophoblast differentiation, it is likely not the only regulatory mechanism involved in this process.
Collapse
Affiliation(s)
- Teena K J B Gamage
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - William Schierding
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Daniel Hurley
- b Systems Biology Laboratory, Melbourne School of Engineering , University of Melbourne , Melbourne , Australia
| | - Peter Tsai
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Jackie L Ludgate
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | | | - Lawrence W Chamley
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Robert J Weeks
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | - Erin C Macaulay
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | - Joanna L James
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| |
Collapse
|
3
|
|
4
|
Jastrząb R, Łomozik L, Tylkowski B. Complexes of biogenic amines in their role in living systems. PHYSICAL SCIENCES REVIEWS 2016. [DOI: 10.1515/psr-2016-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
5
|
Mohammed A, Janakiram NB, Madka V, Ritchie RL, Brewer M, Biddick L, Patlolla JMR, Sadeghi M, Lightfoot S, Steele VE, Rao CV. Eflornithine (DFMO) prevents progression of pancreatic cancer by modulating ornithine decarboxylase signaling. Cancer Prev Res (Phila) 2014; 7:1198-209. [PMID: 25248858 PMCID: PMC4310684 DOI: 10.1158/1940-6207.capr-14-0176] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ornithine decarboxylase (ODC) is the key rate-limiting enzyme in the polyamine synthesis pathway and it is overexpressed in a variety of cancers. We found that polyamine synthesis and modulation of ODC signaling occurs at early stages of pancreatic precursor lesions and increases as the tumor progresses in Kras-activated p48(Cre/+)-LSL-Kras(G12D/+) mice. Interest in use of the ODC inhibitor eflornithine (DFMO) as a cancer chemopreventive agent has increased in recent years since ODC was shown to be transactivated by the c-myc oncogene and to cooperate with the ras oncogene in malignant transformation of epithelial tissues. We tested the effects of DFMO on pancreatic intraepithelial neoplasias (PanIN) and their progression to pancreatic ductal adenocarcinoma (PDAC) in genetically engineered Kras mice. The Kras(G12D/+) mice fed DFMO at 0.1% and 0.2% in the diet showed a significant inhibition (P < 0.0001) of PDAC incidence compared with mice fed control diet. Pancreatic tumor weights were decreased by 31% to 43% (P < 0.03-0.001) with both doses of DFMO. DFMO at 0.1% and 0.2% caused a significant suppression (27% and 31%; P < 0.02-0.004) of PanIN 3 lesions (carcinoma in situ). DFMO-treated pancreas exhibited modulated ODC pathway components along with decreased proliferation and increased expression of p21/p27 as compared with pancreatic tissues derived from mice fed control diet. In summary, our preclinical data indicate that DFMO has potential for chemoprevention of pancreatic cancer and should be evaluated in other PDAC models and in combination with other drugs in anticipation of future clinical trials.
Collapse
Affiliation(s)
- Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Naveena B Janakiram
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rebekah L Ritchie
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Misty Brewer
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Laura Biddick
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jagan Mohan R Patlolla
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael Sadeghi
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Stan Lightfoot
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Vernon E Steele
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Bethesda, Maryland
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hem-Onc Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
6
|
Burnette M, Brito-Robinson T, Li J, Zartman J. An inverse small molecule screen to design a chemically defined medium supporting long-term growth of Drosophila cell lines. MOLECULAR BIOSYSTEMS 2014; 10:2713-23. [PMID: 25096480 PMCID: PMC4890965 DOI: 10.1039/c4mb00155a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drosophila cell culture is used as a model system with multiple applications including the identification of new therapeutic targets in screens, the study of conserved signal transduction pathway mechanisms, and as an expression system for recombinant proteins. However, in vitro methods for Drosophila cell and organ cultures are relatively undeveloped. To characterize the minimal requirements for long-term maintenance of Drosophila cell lines, we developed an inverse screening strategy to identify small molecules and synergies stimulating proliferation in a chemically defined medium. In this chemical-genetics approach, a compound-protein interaction database is used to systematically score genetic targets on a screen-wide scale to extract further information about cell growth. In the pilot screen, we focused on two well-characterized cell lines, Clone 8 (Cl.8) and Schneider 2 (S2). Validated factors were investigated for their ability to maintain cell growth over multiple passages in the chemically defined medium (CDM). The polyamine spermidine proved to be the critical component that enables the CDM to support long-term maintenance of Cl.8 cells. Spermidine supplementation upregulates DNA synthesis for Cl.8 and S2 cells and increases MAPK signaling for Cl.8 cells. The CDM also supports the long-term growth of Kc167 cells. Our target scoring approach validated the importance of polyamines, with enrichment for multiple polyamine ontologies found for both cell lines. Future iterations of the screen will enable the identification of compound combinations optimized for specific applications-maintenance and generation of new cell lines or the production and purification of recombinant proteins-thus increasing the versatility of Drosophila cell culture as both a genetic and biochemical model system. Our cumulative target scoring approach improves on traditional chemical-genetics methods and is extensible to biological processes in other species.
Collapse
Affiliation(s)
- M Burnette
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | | | |
Collapse
|
7
|
Alvarez-Múgica M, Fernández-Gómez JM, Cebrian V, Fresno F, Escaf S, Sánchez-Carbayo M. Polyamine-modulated factor-1 methylation predicts Bacillus Calmette-Guérin response in patients with high-grade non-muscle-invasive bladder carcinoma. Eur Urol 2012; 63:364-70. [PMID: 22682992 DOI: 10.1016/j.eururo.2012.05.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/28/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bacillus Calmette-Guérin (BCG) is a standard treatment to reduce tumor recurrence and delay progression of high-risk non-muscle-invasive (NMI) bladder tumors. However, it is not clear yet which patients are more likely to respond to BCG. OBJECTIVE The aim was to evaluate the role of polyamine-modulated factor-1 (PMF-1) methylation in predicting clinical outcome of T1 high-grade (T1HG) bladder tumors treated with BCG. DESIGN, SETTING, AND PARTICIPANTS In a retrospective design, PMF-1 methylation was analyzed on tumor specimens belonging to 108 patients with T1HG NMI bladder cancer undergoing BCG treatment. Median follow-up was 77 mo (range: 5-235 mo). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS PMF-1 methylation was assessed by methylation-specific polymerase chain reactions. Recurrence, progression into muscle-invasive tumors, and disease-specific survival rates were analyzed using competing risks regression analysis. RESULTS AND LIMITATIONS Among the 108 patients analyzed, 35 had recurring disease (32.4%), 21 progressed (19.4%), and 16 died of disease (14.8%); 71.3% of tumors had PMF-1 methylation. Univariate analyses using cumulative incidence curves revealed that an unmethylated PMF-1 was significantly associated with increased recurrence (p=0.026), progression (p=0.01), and shorter disease-specific survival (log-rank, p=0.03). Multivariate analyses indicated that among sex, age, focality, tumor size, and concomitant carcinoma in situ, only PMF-1 methylation provided significant hazard ratios (HRs) for recurrence of (HR: 2.032; p=0.042), and progression (HR: 2.910; p=0.020). Limitations of the study include its retrospective design, lymphovascular invasion status not available, short maintenance BCG, and that a second transurethral resection was not performed. CONCLUSIONS Epigenetic analyses revealed that the methylation status of PMF-1 was associated with the clinical outcome of patients with T1HG tumors undergoing BCG treatment. An unmethylated PMF-1 correlated to recurrence and progression in T1HG disease using univariate and multivariate analyses. Thus, assessing the methylation status of PMF-1 may serve to distinguish patients responding to BCG from those who may require more aggressive therapeutic approaches.
Collapse
Affiliation(s)
- Miguel Alvarez-Múgica
- Tumor Markers Group, Molecular Pathology Program, Spanish National Cancer Center, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
8
|
Dong Y, Zhu Y, Li J, Zhou QH, Wu C, Oupický D. Synthesis of bisethylnorspermine lipid prodrug as gene delivery vector targeting polyamine metabolism in breast cancer. Mol Pharm 2012; 9:1654-64. [PMID: 22545813 DOI: 10.1021/mp300001m] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Progress in the development of nonviral gene delivery vectors continues to be hampered by low transfection activity and toxicity. Here we proposed to develop a lipid prodrug based on a polyamine analogue bisethylnorspermine (BSP) that can function dually as gene delivery vector and, after intracellular degradation, as active anticancer agent targeting dysregulated polyamine metabolism. We synthesized a prodrug of BSP (LS-BSP) capable of intracellular release of BSP using thiolytically sensitive dithiobenzyl carbamate linker. Biodegradability of LS-BSP contributed to decreased toxicity compared with nondegradable control L-BSP. BSP showed a strong synergistic enhancement of cytotoxic activity of TNF-related apoptosis-inducing ligand (TRAIL) in human breast cancer cells. Decreased enhancement of TRAIL activity was observed for LS-BSP when compared with BSP. LS-BSP formed complexes with plasmid DNA and mediated transfection activity comparable to DOTAP and L-BSP. Our results show that BSP-based vectors are promising candidates for combination drug/gene delivery.
Collapse
Affiliation(s)
- Yanmei Dong
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, Michigan 48202, United States
| | | | | | | | | | | |
Collapse
|
9
|
Bisethylnorspermine lipopolyamine as potential delivery vector for combination drug/gene anticancer therapies. Pharm Res 2010; 27:1927-38. [PMID: 20577786 DOI: 10.1007/s11095-010-0197-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 06/15/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE To design novel synthetic gene delivery system in which the carrier molecule functions dually as a carrier and a cytotoxic agent targeting dysregulated polyamine metabolism in cancer. METHODS Bisethylnorspermine (BENSpm) lipopolyamine was synthesized and its toxicity evaluated by MTS assay in MCF-7 and MCF-10A cells. Transfection activity was determined using luciferase plasmid DNA. RESULTS Asymmetrical lipid analogue of polyamine anticancer drug BENSpm was synthesized using nucleophilic ring opening of azetidinium ion. The synthesized LipoBENSpm showed cytotoxicity comparable to that of parent BENSpm in human breast cancer cell line MCF-7 but mediated 3-4 orders magnitude higher transfection activity. Importantly, cytostatic activity of BENSpm, typically in a low muM range, falls within a relevant and typical concentration range required for efficient gene delivery. CONCLUSIONS These findings make gene delivery vectors based on BENSpm promising candidates for combination drug/gene approaches to the treatment of cancer.
Collapse
|
10
|
Aleman A, Cebrian V, Alvarez M, Lopez V, Orenes E, Lopez-Serra L, Algaba F, Bellmunt J, López-Beltrán A, Gonzalez-Peramato P, Cordon-Cardo C, García J, del Muro JG, Esteller M, Sánchez-Carbayo M. Identification of PMF1 methylation in association with bladder cancer progression. Clin Cancer Res 2009; 14:8236-43. [PMID: 19088041 DOI: 10.1158/1078-0432.ccr-08-0778] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Polyamines are important regulators of cell growth and death. The polyamine modulated factor-1 (PMF-1) is involved in polyamine homeostasis. After identifying an enriched CpG island encompassing the PMF1 promoter, we aimed at evaluating the clinical relevance of PMF1 methylation in bladder cancer. EXPERIMENTAL DESIGN The epigenetic silencing of PMF1 by hypermethylation was tested in bladder cancer cells (n = 11) after azacytidine treatment. PMF1 methylation status was evaluated in 507 bladder tumors and 118 urinary specimens of bladder cancer patients and controls. PMF1 protein expression was analyzed by immunohistochemistry on tissue arrays containing bladder tumors for which PMF1 methylation was assessed (n = 218). RESULTS PMF1 hypermethylation was associated with gene expression loss, being restored in vitro by a demethylating agent. An initial set of 101 primary frozen bladder tumors served to identify PMF1 hypermethylation in 88.1% of the cases. An independent set of 406 paraffin-embedded tumors also revealed a high PMF1 methylation rate (77.6%). PMF1 methylation was significantly associated with increasing stage (P = 0.025). Immunohistochemical analyses revealed that PMF1 methylation was associated with cytoplasmic PMF1 expression loss (P = 0.032). PMF1 protein expression patterns were significantly associated with stage (P < 0.001), grade (P < 0.001), and poor overall survival using univariate (P < 0.001) and multivariate (P = 0.011) analyses. Moreover, PMF1 methylation in urinary specimens distinguished bladder cancer patients from controls (area under the curve = 0.800). CONCLUSION PMF1 was identified to be epigenetically modified in bladder cancer. The association of PMF1 methylation with tumor progression and its diagnostic ability using urinary specimens support including PMF1 assessment for the clinical management of bladder cancer patients.
Collapse
Affiliation(s)
- Ainel Aleman
- Tumor Markers Group, Molecular Pathology Program, Spanish National Cancer Center, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Raul F. Revival of 2-(difluoromethyl)ornithine (DFMO), an inhibitor of polyamine biosynthesis, as a cancer chemopreventive agent. Biochem Soc Trans 2007; 35:353-5. [PMID: 17371277 DOI: 10.1042/bst0350353] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ODC (ornithine decarboxylase), a key enzyme of polyamine biosynthesis, is an inducible enzyme exhibiting high activity in tumour cells, suggesting ODC as a target for antineoplastic therapy. Among the inhibitors of polyamine-related enzymes, the ODC inactivator DFMO [2-(difluoromethyl)ornithine] became the most well-known. The drug is usually cytostatic and its effects on growth are reversed by micromolar concentrations of polyamines in the cellular environment. ODC inactivation is associated with decreased transcription of the growth-related c-myc and c-fos genes. DFMO used as a single drug has only minor effects on tumour growth. The low efficacy of the drug is due to the use of exogenous (gastrointestinal) polyamines by the mammalian organism. Although it was disappointing in most therapeutic attempts, DFMO showed potential in cancer chemoprevention based on its ability to lower polyamine levels in colorectal mucosa at low dosages with no demonstrable toxicity over long periods of use. DFMO in combination with other drugs prevents and inhibits the development of a variety of chemically induced cancers in animals with doses far lower than those administered for therapy. Low doses of several NSAIDs (non-steroidal anti-inflammatory drugs) and DFMO administered in combination have been shown to be more effective in inhibiting chemically induced colon tumours in rats than are high doses of these agents given individually. This combination has gained further interest after findings suggesting that ODC polymorphism is a genetic marker for colon cancer risk and supporting the use of DFMO and aspirin or other NSAIDs in combination as a strategy for colon cancer prevention.
Collapse
Affiliation(s)
- F Raul
- INSERM U682, University Louis Pasteur EA 3430, Laboratory of Nutritional Cancer Prevention, IRCAD, 1 Place de l'Hôpital, 67091 Strasbourg Cedex, France.
| |
Collapse
|
12
|
Casero RA, Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 2007; 6:373-90. [PMID: 17464296 DOI: 10.1038/nrd2243] [Citation(s) in RCA: 570] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The polyamines spermidine and spermine and their diamine precursor putrescine are naturally occurring, polycationic alkylamines that are essential for eukaryotic cell growth. The requirement for and the metabolism of polyamines are frequently dysregulated in cancer and other hyperproliferative diseases, thus making polyamine function and metabolism attractive targets for therapeutic intervention. Recent advances in our understanding of polyamine function, metabolic regulation, and differences between normal cells and tumour cells with respect to polyamine biology, have reinforced the interest in this target-rich pathway for drug development.
Collapse
Affiliation(s)
- Robert A Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
13
|
Babbar N, Ignatenko NA, Casero RA, Gerner EW. Cyclooxygenase-independent induction of apoptosis by sulindac sulfone is mediated by polyamines in colon cancer. J Biol Chem 2003; 278:47762-75. [PMID: 14506281 DOI: 10.1074/jbc.m307265200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Sulindac, a non-steroidal anti-inflammatory prodrug, is metabolized into pharmacologically active sulfide and sulfone derivatives. Sulindac sulfide, but not sulindac sulfone, inhibits cyclooxygenase (COX) enzyme activities, yet both derivatives have growth inhibitory effects on colon cancer cells. Microarray analysis was used to detect COX-independent effects of sulindac on gene expression in human colorectal cells. Spermidine/sperm-ine N1-acetyltransferase (SSAT) gene, which encodes a polyamine catabolic enzyme, was induced by clinically relevant sulindac sulfone concentrations. Northern blots confirmed increased SSAT RNA levels in these colon cancer cells. Deletion analysis and mutational studies were done to map the sulindac sulfone-dependent response sequences in the SSAT 5'-flanking sequences. This led us to the identification of two peroxisome proliferator-activated receptor (PPAR) response elements (PPREs) in the SSAT gene. PPRE-2, at +48 bases relative to the transcription start site, is required for the induction of SSAT by sulindac sulfone and is specifically bound by PPAR gamma in the Caco-2 cells as shown by transfection and gel shift experiments. PPRE-1, at-323 bases relative to the start site, is not required for the induction of SSAT by sulindac sulfone but can be bound by both PPAR delta and PPAR gamma. Sulindac sulfone reduced cellular polyamine contents in the absence but not in the presence of verapamil, an inhibitor of the export of monoacetyl diamines, inhibited cell proliferation and induced apoptosis. The induced apoptosis could be partially rescued by exogenous putrescine. These data suggest that apoptosis induced by sulindac sulfone is mediated, in part, by the COX-independent, PPAR-dependent transcriptional activation of SSAT, leading to reduced tissue polyamine contents in human colon cancer cells.
Collapse
MESH Headings
- Acetyltransferases/genetics
- Apoptosis
- Blotting, Northern
- Blotting, Western
- Caco-2 Cells
- Cell Membrane/metabolism
- Cell Survival
- Colonic Neoplasms/metabolism
- Cyclooxygenase 2
- DNA, Complementary/metabolism
- Dose-Response Relationship, Drug
- Humans
- Immunoblotting
- Isoenzymes/metabolism
- Membrane Proteins
- Models, Biological
- Models, Genetic
- Oligonucleotide Array Sequence Analysis
- Oligonucleotides/chemistry
- Plasmids/metabolism
- Polyamines/chemistry
- Promoter Regions, Genetic
- Prostaglandin-Endoperoxide Synthases/metabolism
- Protein Binding
- Protein Biosynthesis
- Putrescine/chemistry
- RNA/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Response Elements
- Spermidine/metabolism
- Sulindac/analogs & derivatives
- Sulindac/pharmacology
- Time Factors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Naveen Babbar
- Arizona Cancer Center, and Biochemistry, Molecular and Cellular Biology Graduate Program, The University of Arizona, Tucson, 85724, USA
| | | | | | | |
Collapse
|
14
|
Wang Y, Devereux W, Stewart TM, Casero RA. Polyamine-modulated factor 1 binds to the human homologue of the 7a subunit of the Arabidopsis COP9 signalosome: implications in gene expression. Biochem J 2002; 366:79-86. [PMID: 12020345 PMCID: PMC1222765 DOI: 10.1042/bj20020211] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2002] [Revised: 04/16/2002] [Accepted: 05/20/2002] [Indexed: 01/11/2023]
Abstract
Polyamines have been identified to play a role in the transcription of various growth-related genes. The recently discovered polyamine responsive element and the associated trans-acting proteins involved in polyamine-regulated transcription have provided a model system for the study of the role of polyamines in transcription. Polyamine-modulated factor 1 (PMF-1) was identified as one of the transacting factors that binds to NF-E2 related factor-2 (Nrf-2) to regulate the transcription of spermidine/spermine N(1)-acetyltransferase (SSAT). The possibility that PMF-1 also binds to other proteins involved in transcriptional regulation cannot be ruled out. Using a yeast two-hybrid strategy, it was found that PMF-1 binds to a human homologue of the Arabidopsis COP9 signalosome subunit 7a (CSN 7) protein. In the present study, we describe human CSN 7, a 275-amino-acid- containing protein that may have a direct role in regulating gene expression. CSN 7 and PMF-1 bind to each other, as well as compete with each other for binding to Nrf-2. This competition for Nrf-2 binding and interaction with each other is implicated in the regulation of SSAT transcription. CSN 7 possesses a C-terminal coiled-coil domain similar to the domain that mediates the interaction between PMF-1 and Nrf-2, suggesting that coiled-coil domains also mediate the interaction between CSN 7 and PMF-1. Since CSN 7 does not contain a DNA-binding domain, its effects on transcription must occur in conjunction with binding to other proteins. The results presented here demonstrate that PMF-1 and Nrf-2 can act as protein partners of CSN 7.
Collapse
Affiliation(s)
- Yanlin Wang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Oncology Center, Bunting-Blaustein Cancer Research Building, Room 551, 1650 Orleans Street, Baltimore, MD 21231, U.S.A
| | | | | | | |
Collapse
|
15
|
Lin X, Tascilar M, Lee WH, Vles WJ, Lee BH, Veeraswamy R, Asgari K, Freije D, van Rees B, Gage WR, Bova GS, Isaacs WB, Brooks JD, DeWeese TL, De Marzo AM, Nelson WG. GSTP1 CpG island hypermethylation is responsible for the absence of GSTP1 expression in human prostate cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:1815-26. [PMID: 11696442 PMCID: PMC1867052 DOI: 10.1016/s0002-9440(10)63028-3] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
GSTP1 CpG island hypermethylation is the most common somatic genome alteration described for human prostate cancer (PCA); lack of GSTP1 expression is characteristic of human PCA cells in vivo. We report here that loss of GSTP1 function may have been selected during the pathogenesis of human PCA. Using a variety of techniques to detect GSTP1 CpG island DNA hypermethylation in PCA DNA, we found only hypermethylated GSTP1 alleles in each PCA cell in all but two PCA cases studied. In these two cases, CpG island hypermethylation was present at only one of two GSTP1 alleles in PCA DNA. In one of the cases, DNA hypermethylation at one GSTP1 allele and deletion of the other GSTP1 allele were evident. In the other case, an unmethylated GSTP1 allele was detected, accompanied by abundant GSTP1 expression. GSTP1 CpG island DNA hypermethylation was responsible for lack of GSTP1 expression by LNCaP PCA cells: treatment of the cells with 5-azacytidine (5-aza-C), an inhibitor of DNA methyltransferases, reversed the GSTP1 promoter DNA hypermethylation, activated GSTP1 transcription, and restored GSTP1 expression. GSTP1 promoter activity, assessed via transfection of GSTP1 promoter-CAT reporter constructs in LNCaP cells, was inhibited by SssI-catalyzed CpG dinucleotide methylation. Remarkably, although selection for loss of GSTP1 function may be inferred for human PCA, GSTP1 did not act like a tumor suppressor gene, as LNCaP cells expressing GSTP1, either after 5-aza-C treatment or as a consequence of transfection with GSTP1 cDNA, grew well in vitro and in vivo. Perhaps, GSTP1 inactivation may render prostatic cells susceptible to additional genome alterations, caused by electrophilic or oxidant carcinogens, that provide a selective growth advantage.
Collapse
Affiliation(s)
- X Lin
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang Y, Devereux W, Woster PM, Casero RA. Cloning and characterization of the mouse polyamine-modulated factor-1 (mPMF-1) gene: an alternatively spliced homologue of the human transcription factor. Biochem J 2001; 359:387-92. [PMID: 11583586 PMCID: PMC1222158 DOI: 10.1042/0264-6021:3590387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The natural polyamines and their analogues have been implicated in transcriptional regulation of specific genes. Human polyamine-modulated factor-1 (hPMF-1) was the first polyamine-responsive transcription factor identified. Here the mouse homologue of the hPMF-1 gene is described. Interestingly, the mouse gene (mPMF-1) codes for two alternatively spliced mRNAs. Both of the mouse splice variants, mPMF-1S and mPMF-1L, possess C-terminal coiled-coil domains nearly identical to that found in hPMF-1 and are highly homologous with the human protein. The C-terminal coiled-coil structure is necessary for transcriptional activation. However, the shorter protein, mPMF-1S, does not contain an N-terminal coiled-coil region as do both hPMF-1 and the longer mPMF-1L. mPMF-1L mRNA codes for a protein of 202 amino acids, 37 amino acids longer than the human protein. By contrast, mPMF-1S codes for only 133 amino acids, as a result of two exons being omitted compared with mPMF-1L. Both mouse transcription factors can interact with Nrf-2 (nuclear factor-E2-related factor 2), the normal partner of hPMF-1, substantiating the importance of the C-terminal coiled-coil region responsible for this interaction. Finally, the expression of mPMF-1 is induced when mouse M1 myeloid leukaemia cells are exposed to polyamine analogues, suggesting control similar to that observed for the hPMF-1.
Collapse
Affiliation(s)
- Y Wang
- The Johns Hopkins Oncology Center, Bunting Blaustein Cancer Research Building, Room 551, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | |
Collapse
|
17
|
Tabib A, Bachrach U. Role of polyamines in mediating malignant transformation and oncogene expression. Int J Biochem Cell Biol 1999; 31:1289-95. [PMID: 10605821 DOI: 10.1016/s1357-2725(99)00098-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Previous studies have demonstrated that polyamines accumulate in cancer cells and that overproduction of ornithine decarboxylase (ODC), which catalyzes polyamine synthesis, elicits the acquisition of the transformed phenotype. However, it was not clear whether the expression of ODC and the accumulation of polyamines are only innocent by-products of the transformation process. In this study we confirm previous findings what polyamines can trigger the transformation of immortalized cultured cells. In addition to NIH 3T3 fibroblasts, studied previously, rat kidney epithelial cells or fibroblasts also grew in soft agar in the presence of polyamines. It has also been demonstrated that spermidine, preferentially stimulated the transcription and the expression of c-myc while those of c-fos were preferentially stimulated by putrescine. These findings suggest that the effect of polyamines on cellular transformation, could be explained, at least partially, by stimulation of proto-oncogene expression.
Collapse
Affiliation(s)
- A Tabib
- Department of Molecular Biology, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
18
|
Manteuffel-Cymborowska M, Peska M, Chmurzyńska W, Grzelakowska-Sztabert B. Catecholamines are required for androgen-induced ODC expression but not for hypertrophy of mouse kidney. BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1356:292-8. [PMID: 9194572 DOI: 10.1016/s0167-4889(97)00011-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Catecholamine depletion, evoked by reserpine, dramatically impaired (5-fold) the testosterone-induced increase of ornithine decarboxylase (ODC) activity in female mouse kidney. However, reserpine did not prevent kidney hypertrophy evoked by testosterone. This is evidenced by the activity of sensitive, biochemical markers of renal hypertrophy, namely arginase and ornithine aminotransferase (OAT), that responded with the increase and decrease of activities to testosterone treatment, respectively. Arginine and ornithine, substrates and/or products of marker enzymes, showed a striking homeostasis as their level was not affected by testosterone and reserpine, and only slightly by DFMO. Northern blot analysis revealed that the ODC mRNA level, that was increased 10-fold by testosterone, was decreased 2-fold in catecholamine-depleted hypertrophic kidney. Thus, ODC transcript level, lowered by reserpine, correlated partially with an attenuated response of ODC activity to testosterone. This was in contrast to DFMO, which inhibited ODC activity, but significantly increased its mRNA content. It is concluded that catecholamines could be involved together with testosterone in regulation of the ODC gene expression in mouse kidney.
Collapse
|
19
|
Blachier F, Selamnia M, Robert V, M'Rabet-Touil H, Duée PH. Metabolism of L-arginine through polyamine and nitric oxide synthase pathways in proliferative or differentiated human colon carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1268:255-62. [PMID: 7548223 DOI: 10.1016/0167-4889(95)00083-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
HT-29 Glc-/+ cells originate from a human colon adenocarcinoma. These cells have been selected in a glucose-free culture medium and switched back in a glucose-containing medium. In this condition, they can spontaneously differentiate after confluency in enterocyte-like cells according to the activity of the brush-border associated hydrolase dipeptidyl peptidase IV. Since L-arginine can generate polyamines which are necessary for cellular proliferation and also differentiation, and nitric oxide with reported anti-proliferative property, the metabolism of this amino acid was examined in proliferative and differentiated isolated HT-29 cells. Proliferative HT-29 cells were characterized by micromolar intracellular concentration of putrescine and millimolar concentration of spermidine and spermine. In these cells, L-arginine is converted to L-ornithine and putrescine and to a minor part to nitric oxide and L-citrulline. Putrescine was taken up by HT-29 cells, leading to the production of a modest amount of spermidine. The diamine was slightly incorporated into cellular proteins and largely released in the incubation medium. The proliferative HT-29 cells take up spermidine and spermine but do not catabolize these polyamines and slightly released spermidine. Differentiation of HT-29 cells is not associated with change in intracellular polyamine content but is paralleled by an almost complete extinction of de novo synthesis of putrescine (due to a dramatic decrease of ornithine decarboxylase activity) and by a reduced release capacity of putrescine. In contrast, putrescine net uptake and incorporation into cellular proteins remained unchanged after differentiation. Furthermore, spermidine and spermine metabolism as well as the circulation of L-arginine in the nitric oxide synthase pathway were also not modified after differentiation. In conclusion, putrescine is the L-arginine-derived molecule, the metabolism of which is specifically and markedly modified when HT-29 cells move from proliferative to differentiated state.
Collapse
Affiliation(s)
- F Blachier
- Unité d'Ecologie et de Physiologie du Système Digestif. Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | | | | | | | | |
Collapse
|
20
|
Clément S, Delcros JG, Basu HS, Quash G, Marton LJ, Feuerstein BG. The structure of polyamine analogues determines haemoglobin production and cytotoxicity in murine erythroleukaemia cells. Biochem J 1995; 309 ( Pt 3):787-91. [PMID: 7639694 PMCID: PMC1135701 DOI: 10.1042/bj3090787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The naturally occurring polyamine spermine induces haemoglobin synthesis in murine erythroleukaemia (MEL) cells. We have studied the ability of various polyamine analogues to inhibit cell growth and induce haemoglobin production. Polyamine analogues with free terminal amino groups were good inducers of haemoglobin production in MEL cells. Haemoglobin levels correlated with the number of positive charges: pentamines (five positive charges) were stronger inducers than tetramines (four positive charges). Compounds ethylated at their terminal amines were poor inducers of haemoglobin production but good inhibitors of MEL cell growth. These results provide evidence that polyamine analogues support specific biological functions of polyamines in MEL cells and suggest relationships between polyamine structure and function.
Collapse
Affiliation(s)
- S Clément
- Laboratoire d'Immunochimie INSERM C.J.F. 89-05, Faculté de Médecine Lyon Sud, Oullins, France
| | | | | | | | | | | |
Collapse
|
21
|
Wallon UM, Shassetz LR, Cress AE, Bowden GT, Gerner EW. Polyamine-dependent expression of the matrix metalloproteinase matrilysin in a human colon cancer-derived cell line. Mol Carcinog 1994; 11:138-44. [PMID: 7945802 DOI: 10.1002/mc.2940110304] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Matrilysin, which is a member of the matrix metalloproteinase family and is implicated in colon cancer invasion, is expressed in human colon adenocarcinoma-derived SW1116 cells. We investigated the effect of alpha-difluoromethylornithine (DFMO) on matrilysin expression in this cell line because others have shown that DFMO can inhibit invasion and carcinogenesis in epithelial tissues, including the colon, in experimental models. DFMO reduced extracellular levels of matrilysin protein after 4 d of treatment. Intracellular levels of matrilysin protein were minimally affected by DFMO treatment. The decrease in extracellular matrilysin protein levels caused by DFMO was not a consequence of lowered steady-state levels of matrilysin mRNA. After 4 d of exposure, the amount of this transcript was higher in DFMO-treated cells than in untreated cultures, whereas the mRNA stabilities were similar. These data show that polyamine depletion by DFMO can suppress the expression of matrilysin, a gene product thought to be involved in tumor invasion. The decrease in extracellular matrilysin protein caused by DFMO treatment appears to be due to a posttranscriptional mechanism, although transcription of this gene also seems to be affected by polyamines in SW1116 cells.
Collapse
Affiliation(s)
- U M Wallon
- Department of Radiation Oncology, University of Arizona Health Sciences Center, Tucson 85724
| | | | | | | | | |
Collapse
|
22
|
Fogel-Petrovic M, Kramer DL, Ganis B, Casero RA, Porter CW. Cloning and sequence analysis of the gene and cDNA encoding mouse spermidine/spermine N1-acetyltransferase--a gene uniquely regulated by polyamines and their analogs. BIOCHIMICA ET BIOPHYSICA ACTA 1993; 1216:255-64. [PMID: 8241266 DOI: 10.1016/0167-4781(93)90152-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The polyamine catabolizing enzyme, spermidine/spermine N1-acetyltransferase (SSAT), has been implicated as a critical determinant of polyamine pool maintenance. SSAT has recently been shown to be positively regulated in human cell lines by polyamines and their analogs at the level of mRNA accumulation. Mouse LA-4 lung adenoma cells treated with either spermine or the spermine analog, N1,N12-bis(ethyl)spermine, produced a 2.3 and 6.5-fold increase, respectively, in SSAT mRNA. Prior evidence for transcriptional control of the enzyme prompted investigation of SSAT gene structure and its regulatory elements. The mouse SSAT gene was isolated as a 3650 bp EcoRI fragment from a lambda-J1 Mus saxicola genomic library by hybridization with human SSAT cDNA. An additional 431 bp downstream from the 3' EcoRI site were sequenced from a BamHI fragment (total gene sequence, 4066 bp). The gene contains six exons and five introns. Sequence analysis of the 774 bp of the 5' non-coding region revealed the absence of TATAA or CCAAT sequence motifs and the presence of a number of binding motifs in the 5' region of the gene with consensus binding sequences for transcription factors SP1, AP1, E2F, AP2, PEA-3 and others. The deduced amino acid sequence of the coding region differs from that of the human SSAT cDNA by five amino acids. The 527 bp of the 3' non-coding region contains four possible polyadenylation signal sites of which only one displays a typical consensus sequence. A 940 bp SSAT cDNA was isolated from Mus domesticus (BALB-C) liver lambda gt11 cDNA library. It contains a 5' untranslated region 89 bp in length and a 3' untranslated region 376 bp in length. The amino acid sequence deduced from Mus domesticus differs from that of Mus saxicola by one amino acid, from the hamster cDNA, by four amino acids and from the human cDNA by six amino acids. Further elucidation of the structural features of the SSAT gene may reveal how it is positively regulated by polyamines and their analogs.
Collapse
Affiliation(s)
- M Fogel-Petrovic
- Grace Cancer Drug Center, Roswell Park Cancer Institute, Buffalo, NY 14263-0001
| | | | | | | | | |
Collapse
|
23
|
Fogel-Petrovic M, Shappell N, Bergeron R, Porter C. Polyamine and polyamine analog regulation of spermidine/spermine N1-acetyltransferase in MALME-3M human melanoma cells. J Biol Chem 1993. [DOI: 10.1016/s0021-9258(17)46742-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
24
|
Shappell NW, Fogel-Petrovic MF, Porter CW. Regulation of spermidine/spermine N1-acetyltransferase by intracellular polyamine pools. Evidence for a functional role in polyamine homeostasis. FEBS Lett 1993; 321:179-83. [PMID: 8477847 DOI: 10.1016/0014-5793(93)80103-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Through its role in polyamine acetylation and the back-conversion pathway, spermidine/spermine N1-acetyltransferase (SSAT) has the potential to control intracellular polyamine pools by facilitating their catabolism and/or excretion. The possibility that the enzyme is subject to regulation by intracellular polyamine pools was investigated in MALME-3 human melanoma cells. Increases in intracellular polyamine pools by treatment with 3 microM exogenous spermidine or spermine for 48 h caused SSAT activity to increase 111% and 226%, respectively, and SSAT-specific mRNA to rise 19% and 66%, respectively. Decreases in polyamine pools by treatment with inhibitors of polyamine biosynthesis caused SSAT activity to decrease by 46% and mRNA to fall by 89%. Both SSAT activity and mRNA were more sensitive to changes in spermine than spermidine. The identification of a positive regulatory relationship between SSAT and intracellular polyamine pools further implicates this enzyme in a proposed model for polyamine pool homeostasis.
Collapse
Affiliation(s)
- N W Shappell
- Grace Cancer Drug Center, Roswell Park Cancer Institute, Buffalo, NY 14263-0001
| | | | | |
Collapse
|
25
|
Wang H, Scott RE. Induction of c-jun independent of PKC, pertussis toxin-sensitive G protein, and polyamines in quiescent SV40-transformed 3T3 T cells. Exp Cell Res 1992; 203:47-55. [PMID: 1330658 DOI: 10.1016/0014-4827(92)90038-a] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CSV3 clones of simian virus 40 large T antigen-transformed murine 3T3 T cells can be made quiescent as part of a differentiation process. In these quiescent cells, insulin- and vanadate-induced mitogenesis are both associated with the induction of the c-jun proto-oncogene (Wang and Scott 1991 J. Cell. Physiol. 147, 102-110; Wang et al. 1991 Cell Growth Differ. 2, 645-652). The current studies were therefore designed to compare the early signal transduction pathways employed by insulin and vanadate to regulate c-jun expression. In quiescent CSV3-1 cells, down-regulation of protein kinase C by prolonged exposure to 12-O-tetra-decanoylphorbol-13-acetate or inhibition of protein kinase C activity by treatment with the protein kinase C antagonist staurosporine is shown not to affect c-jun induction by insulin or vanadate. This suggests that both insulin and vanadate act in a protein kinase C-independent manner. Insulin's effect on c-jun induction does, however, involve a G protein because insulin's effect can be inhibited by pertussis toxin. In contrast, vanadate induction of c-jun is not affected by pertussis toxin. Genistein, a general tyrosine kinase inhibitor, can inhibit the ability of vanadate to induce c-jun but it does not inhibit insulin's effect. Finally, the depletion of polyamines, particularly spermidine, by DL-alpha-difluoromethylornithine treatment also prevents c-jun induction by insulin but DL-alpha-difluoromethylornithine treatment has no effect on c-jun induction by vanadate. These observations indicate that the c-jun induction by insulin and vanadate in CSV3-1 cells is mediated by different signal transduction mechanisms. Together with our previously published data, these results suggest that c-jun can be induced independent of protein kinase C activation, without involvement of pertussis toxin-sensitive G protein, independent of induction of c-fos, and without expression of high levels of intracellular polyamines.
Collapse
Affiliation(s)
- H Wang
- Department of Pathology, University of Tennessee Medical Center, Memphis 38163
| | | |
Collapse
|
26
|
Parekh D, Saydjari R, Ishizuka J, Townsend CM, Thompson JC. Sodium butyrate stimulates polyamine biosynthesis in colon cancer cells. Surg Oncol 1992; 1:315-22. [PMID: 1341266 DOI: 10.1016/0960-7404(92)90093-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Differentiation inducers act through polyamine-dependent and independent pathways. Sodium butyrate (NaB) inhibits proliferation and induces terminal differentiation in human and murine cancer cell lines. An effect of this agent on polyamine biosynthesis has not been demonstrated previously. In the present study, we examined the effects of NaB on polyamine biosynthesis in mouse colon cancer (MC-26) cells. All studies were performed on exponentially growing cells, and ODC and polyamine transport measurements were performed as described previously. NaB inhibited the growth of MC-26 cells in a dose-dependent manner. Cell shape was significantly altered by treatment with NaB (development of dendritic-like processes and flattening and spreading out of cells on culture dishes). NaB stimulated ODC activity in a dose-dependent manner. The activity was elevated by 8 h after treatment, and at 48 h there was a ten-fold increase in activity (compared with control activity). The increase in ODC activity led to an increase in polyamine biosynthesis; putrescine, spermidine, and spermine levels in MC-26 cells were significantly elevated by 24 h after treatment with NaB. Polyamine uptake was similar in control cells and cells treated with NaB alone. Our finding of significant stimulation of polyamine uptake by NaB after inhibition of endogenous synthesis (by an ODC-dependent pathway) in DFMO-treated cells suggests that cellular requirements are increased for polyamines in NaB-treated cells. We conclude that polyamine-dependent processes are important in the mechanism of action of NaB in colon cancer cells.
Collapse
Affiliation(s)
- D Parekh
- Department of Surgery, University of Texas Medical Branch, Galveston 77555-0527
| | | | | | | | | |
Collapse
|
27
|
Celano P, Berchtold C, Kizer D, Weeraratna A, Nelkin B, Baylin S, Casero RA. Characterization of an endogenous RNA transcript with homology to the antisense strand of the human c-myc gene. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(18)42150-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
28
|
Abstract
Spermine, a polyamine, is known to enhance motor functional recovery after a sciatic nerve lesion in the rat. The effect of spermine on the sensory axonal elongation after a sciatic crush was studied with the pinch-test from the sural nerve in the rat. The effect of spermidine, another polyamine, on the motor functional recovery after a trauma was studied by using the toe-spreading ability as an indicator of motor recovery after a sciatic crush in the rat. Spermine enhanced the rate of regeneration of the sensory axons by 16%. Spermidine enhanced the rate of the motor recovery by 30%. These results suggest that not only spermine but also spermidine enhance regeneration of peripheral somatic nerves.
Collapse
Affiliation(s)
- T Kauppila
- Department of Physiology, University of Helsinki, Finland
| |
Collapse
|