1
|
Wei L, Deng C, Zhang B, Wang G, Meng Y, Qin H. SP4 Facilitates Esophageal Squamous Cell Carcinoma Progression by Activating PHF14 Transcription and Wnt/Β-Catenin Signaling. Mol Cancer Res 2024; 22:55-69. [PMID: 37768180 PMCID: PMC10758695 DOI: 10.1158/1541-7786.mcr-22-0835] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 06/13/2023] [Accepted: 09/25/2023] [Indexed: 09/29/2023]
Abstract
Specificity protein 4 transcription factor (SP4), a member of the Sp/Krüppel-like family (KLF), could bind to GT and GC box promoters, and plays an essential role in transcriptional activating. Despite SP4 having been detected to be highly expressed in a variety of human tumors, its biological effect and underlying molecular mechanism in esophageal squamous cell carcinoma (ESCC) remains unclear. Our research discovered that high SP4 expression is detected in primary ESCC specimens and cell lines and is strongly associated with the ESCC tumor grade and poor prognosis. In vitro, knockdown of SP4 suppressed cell proliferation and cell-cycle progression and promoted apoptosis, whereas overexpression of SP4 did the opposite. In vivo, inhibiting SP4 expression in ESCC cells suppresses tumor growth. Subsequently, we demonstrated that SP4 acts as the transcriptional upstream of PHF14, which binds to PHF14 promoter region, thus promoting PHF14 transcription. PHF14 was also significantly expressed in patient tissues and various ESCC cell lines and its expression promoted cell proliferation and inhibited apoptosis. Moreover, knockdown of SP4 inhibited the Wnt/β-catenin signaling pathway, whereas overexpression of PHF14 eliminated the effects of SP4 knockdown in ESCC cells. These results demonstrate that SP4 activates the Wnt/β-catenin signaling pathway by driving PHF14 transcription, thereby promoting ESCC progression, which indicates that SP4 might act as a prospective prognostic indicator or therapeutic target for patients with ESCC. IMPLICATIONS This study identified SP4/PH14 axis as a new mechanism to promote the progression of ESCC, which may serve as a novel therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Li Wei
- Department of Surgery and Anesthesia, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chaowei Deng
- Department of Cell Biology and Genetics/Institute of Genetics and Developmental Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Bo Zhang
- Department of Peripheral Vascular Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Guanghui Wang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yan Meng
- Department of Peripheral Vascular Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hao Qin
- Department of Peripheral Vascular Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
2
|
Swift ML, Sell C, Azizkhan-Clifford J. DNA damage-induced degradation of Sp1 promotes cellular senescence. GeroScience 2021; 44:683-698. [PMID: 34550526 PMCID: PMC9135943 DOI: 10.1007/s11357-021-00456-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/07/2021] [Indexed: 11/28/2022] Open
Abstract
Persistent DNA damage (genotoxic stress) triggers signaling cascades that drive cells into apoptosis or senescence to avoid replicating a damaged genome. Sp1 has been found to play a role in double strand break (DSB) repair, and a link between Sp1 and aging has also been established, where Sp1 protein, but not RNA, levels decrease with age. Interestingly, inhibition ATM reverses the age-related degradation of Sp1, suggesting that DNA damage signaling is involved in senescence-related degradation of Sp1. Proteasomal degradation of Sp1 in senescent cells is mediated via sumoylation, where sumoylation of Sp1 on lysine 16 is increased in senescent cells. Taking into consideration our previous findings that Sp1 is phosphorylated by ATM in response to DNA damage and that proteasomal degradation of Sp1 at DSBs is also mediated by its sumoylation and subsequent interaction with RNF4, we investigated the potential contribution of Sp1’s role as a DSB repair factor in mediating cellular senescence. We report here that Sp1 expression is decreased with a concomitant increase in senescence markers in response to DNA damage. Mutation of Sp1 at serine 101 to create an ATM phospho-null mutant, or mutation of lysine 16 to create a sumo-null mutant, prevents the sumoylation and subsequent proteasomal degradation of Sp1 and results in a decrease in senescence. Conversely, depletion of Sp1 or mutation of Sp1 to create an ATM phosphomimetic results in premature degradation of Sp1 and an increase in senescence markers. These data link a loss of genomic stability with senescence through the action of a DNA damage repair factor.
Collapse
Affiliation(s)
- Michelle L Swift
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, 245 N 15th Street, MS497, Philadelphia, PA, 19102, USA
| | - Christian Sell
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, 245 N 15th Street, MS497, Philadelphia, PA, 19102, USA
| | - Jane Azizkhan-Clifford
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, 245 N 15th Street, MS497, Philadelphia, PA, 19102, USA.
| |
Collapse
|
3
|
Watanabe N, Nakano M, Mitsuishi Y, Hara N, Mano T, Iwata A, Murayama S, Suzuki T, Ikeuchi T, Nishimura M. Transcriptional downregulation of FAM3C/ILEI in the Alzheimer's brain. Hum Mol Genet 2021; 31:122-132. [PMID: 34378027 DOI: 10.1093/hmg/ddab226] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 11/14/2022] Open
Abstract
Amyloid-β (Aβ) accumulation in the brain triggers the pathogenic cascade for Alzheimer's disease (AD) development. The secretory protein FAM3C (also named ILEI) is a candidate for an endogenous suppressor of Aβ production. In this study, we found that FAM3C expression was transcriptionally downregulated in the AD brain. To determine the transcriptional mechanism of the human FAM3C gene, we delineated the minimal 5'-flanking sequence required for basal promoter activity. From a database search for DNA-binding motifs, expression analysis using cultured cells, and promoter DNA-binding assays, we identified SP1 and EBF1 as candidate basal transcription factors for FAM3C, and found that SMAD1 was a putative inducible transcription factor and KLF6 was a transcription repressor for FAM3C. Genomic deletion of the basal promoter sequence from HEK293 and Neuro-2a cells markedly reduced endogenous expression of FAM3C and abrogated SP1- or EBF1-mediated induction of FAM3C. Nuclear protein extracts from AD brains contained lower levels of SP1 and EBF1 than did those from control brains, although the relative mRNA levels of these factors did not differ significantly between the groups. Additionally, the ability of nuclear SP1 and EBF1 in AD brains to bind with the basal promoter sequence-containing DNA probe was reduced compared with the binding ability of these factors in control brains. Thus, the transcriptional downregulation of FAM3C in the AD brain is attributable to the reduced nuclear levels and genomic DNA binding of SP1 and EBF1. An expressional decline in FAM3C may be a risk factor for Aβ accumulation and eventually AD development.
Collapse
Affiliation(s)
- Naoki Watanabe
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Masaki Nakano
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Yachiyo Mitsuishi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Tatsuo Mano
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Atsushi Iwata
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.,Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Toshiharu Suzuki
- Laboratory of Neuroscience, Graduate School of Pharmaceutical Sciences, Hokkaido University, Hokkaido 060-0812, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| |
Collapse
|
4
|
Kang D, Zuo W, Wu Q, Zhu Q, Liu P. Inhibition of Specificity Protein 1 Is Involved in Phloretin-Induced Suppression of Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1358674. [PMID: 32851058 PMCID: PMC7439178 DOI: 10.1155/2020/1358674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/03/2023]
Abstract
Phloretin is a flavonoid existed in various plants and has been reported to possess anticarcinogenic activity. However, the anticancer mechanism of phloretin in prostate cancer (PCa) remains unclear. Here, our in vitro and in vivo experimental data demonstrate that phloretin inhibits the phosphorylation and the activation of EGFR and then inhibits its downstream PI3K/AKT and MEK/ERK1/2 pathways in PCa cells. Inhibition of these two pathways further decreases expression of Sp1 by inhibiting Sp1 gene transcription, induces degradation of Sp1 protein by inhibiting GSK3β phosphorylation, suppresses nucleolin-enhanced translation of Sp1 mRNA by inhibiting nucleolin phosphorylation, and directly inactivates transcription activity of Sp1. Inhibition of Sp1 subsequently decreases the expression of Sp3/4, VEGF, and Survivin and then upregulates apoptosis-related proteins and downregulates cell cycle-related proteins in PCa cells. Finally, phloretin treatment in PCa cells induces cell growth inhibition and apoptosis, suggesting that phloretin may be an effective therapy compound in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Dan Kang
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Wenren Zuo
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Qingxin Wu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Qingyi Zhu
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Ping Liu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
5
|
Priya Dharshini LC, Vishnupriya S, Sakthivel KM, Rasmi RR. Oxidative stress responsive transcription factors in cellular signalling transduction mechanisms. Cell Signal 2020; 72:109670. [PMID: 32418887 DOI: 10.1016/j.cellsig.2020.109670] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 11/18/2022]
Abstract
Oxidative stress results from the imbalances in the development of reactive oxygen species (ROS) and antioxidants defence system resulting in tissue injury. A key issue resulting in the modulation of ROS is that it alters hosts molecular, structural and functional properties which is accomplished via various signalling pathways which either activate or inhibit numerous transcription factors (TFs). Some of the regulators include Nuclear erythroid-2 related factors (Nrf-2), CCAAT/enhancer-binding protein delta (CEBPD), Activator Protein-1 (AP-1), Hypoxia-inducible factor 1(HIF-1), Nuclear factor κB (NF-κB), Specificity Protein-1 (SP-1) and Forkhead Box class O (FoxO) transcription factors. The expression of these transcription factors are dependent upon the stress signal and are sometimes interlinked. They are highly specific having their own regulation cellular events. Depending upon the transcription factors and better knowledge on the type of the oxidative stress help researchers develop safe, novel targets which can serve as efficient therapeutic targets for several disease conditions.
Collapse
Affiliation(s)
| | - Selvaraj Vishnupriya
- Department of Biotechnology, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India
| | - Kunnathur Murugesan Sakthivel
- Department of Biochemistry, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India
| | - Rajan Radha Rasmi
- Department of Biotechnology, PSG College of Arts & Science, Civil Aerodrome Post, Coimbatore, Tamil Nadu 641 014, India.
| |
Collapse
|
6
|
Bajpai R, Nagaraju GP. Specificity protein 1: Its role in colorectal cancer progression and metastasis. Crit Rev Oncol Hematol 2017; 113:1-7. [PMID: 28427500 DOI: 10.1016/j.critrevonc.2017.02.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Indexed: 01/20/2023] Open
Abstract
Specificity protein 1 (Sp1) is a widely expressed transcription factor that plays an important role in the promotion of oncogenes required for tumor survival, progression and metastasis. Sp1 is highly expressed in several cancers including colorectal cancer (CRC) and is related to poor prognosis. Therefore, targeting Sp1 is a rational for CRC therapy. In this review, we will recapitulate the current understanding of Sp1 signaling, its molecular mechanisms, and its potential involvement in CRC growth, progression and metastasis. We will also discuss the current therapeutic drugs for CRC and their mechanism of action via Sp1.
Collapse
Affiliation(s)
- Richa Bajpai
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
7
|
Safe S, Kasiappan R. Natural Products as Mechanism-based Anticancer Agents: Sp Transcription Factors as Targets. Phytother Res 2016; 30:1723-1732. [PMID: 27384261 DOI: 10.1002/ptr.5669] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/16/2016] [Accepted: 06/01/2016] [Indexed: 08/29/2023]
Abstract
Naturally occurring anticancer agents and their derivatives act on multiple pathways to inhibit carcinogenesis and their inhibition of migration, invasion, growth, survival, and metastasis is associated with downregulation of genes associated with these responses. Several phytochemical-derived anticancer drugs including curcumin, betulinic acid, phenethylisothiocyanate and celastrol, and many others induce reactive oxygen species, and their effects on gene regulation show some overlap in various cancer cell lines. We hypothesize that reactive oxygen species-inducing anticancer agents and many other natural products target a common pathway in cancer cells, which initially involves downregulation of specificity protein 1 (Sp1), Sp3, and Sp4, which are highly expressed in tumors/cell lines derived from solid tumors. This hypothesis is supported by several published reports showing that a large number of phytochemical-derived anticancer agents downregulate Sp1, Sp3, Sp4, and pro-oncogenic Sp-regulated genes involved in cell growth (cyclin D1 and growth factor receptors), survival (bcl-2 and survivin), angiogenesis and migration (MMP-9, vascular endothelial growth factor and its receptors), and inflammation (NF-kB). The contribution of this pathway to the anticancer activity of drugs such as curcumin, celastrol, betulinic acid, and phenethylisothiocyanate must be determined in order to optimize clinical applications of drug combinations containing these compounds. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA.
| | - Ravi Kasiappan
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA
| |
Collapse
|
8
|
Hedrick E, Cheng Y, Jin UH, Kim K, Safe S. Specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 are non-oncogene addiction genes in cancer cells. Oncotarget 2016; 7:22245-22256. [PMID: 26967243 PMCID: PMC5008359 DOI: 10.18632/oncotarget.7925] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/23/2016] [Indexed: 12/31/2022] Open
Abstract
Specificity protein (Sp) transcription factor (TF) Sp1 is overexpressed in multiple tumors and is a negative prognostic factor for patient survival. Sp1 and also Sp3 and Sp4 are highly expressed in cancer cells and in this study, we have used results of RNA interference (RNAi) to show that the three TFs individually play a role in the growth, survival and migration/invasion of breast, kidney, pancreatic, lung and colon cancer cell lines. Moreover, tumor growth in athymic nude mice bearing L3.6pL pancreatic cancer cells as xenografts were significantly decreased in cells depleted for Sp1, Sp3 and Sp4 (combined) or Sp1 alone. Ingenuity Pathway Analysis (IPA) of changes in gene expression in Panc1 pancreatic cancer cells after individual knockdown of Sp1, Sp3 and Sp4 demonstrates that these TFs regulate genes and pathways that correlated with the functional responses observed after knockdown but also some genes and pathways that inversely correlated with the functional responses. However, causal IPA analysis which integrates all pathway-dependent changes in all genes strongly predicted that Sp1-, Sp3- and Sp4-regulated genes were associated with the pro-oncogenic activity. These functional and genomic results coupled with overexpression of Sp transcription factors in tumor vs. non-tumor tissues and decreased Sp1 expression with age indicate that Sp1, Sp3 and Sp4 are non-oncogene addiction (NOA) genes and are attractive drug targets for individual and combined cancer chemotherapies.
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Kyounghyun Kim
- Environmental Health, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
9
|
Transcription factor Sp1 prevents TRF2(ΔBΔM)-induced premature senescence in human diploid fibroblasts. Mol Cell Biochem 2016; 414:201-8. [PMID: 26906205 DOI: 10.1007/s11010-016-2672-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 02/17/2016] [Indexed: 01/08/2023]
Abstract
Telomere uncapping is thought to be the fundamental cause of replicative cellular senescence, but the cellular machineries mediating this process have not been fully understood. In the present study, we present the role of Sp1 transcription factor in the state of telomere uncapping using the TRF2(ΔBΔM)-induced senescence model in human diploid fibroblasts. We observed that the expression of Sp1 is down-regulated in the TRF2(ΔBΔM)-induced senescence, which was mediated by ATM and p38 MAPK. In addition, overexpression of Sp1 prevented the TRF2(ΔBΔM)-induced senescence. Among transcriptional targets of Sp1, expression levels of nuclear transport genes such as karyopherin α, Nup107, and Nup50 were down-regulated in the TRF2(ΔBΔM)-induced senescence, which was prevented by Sp1 overexpression. Moreover, inhibition of the nuclear transport by wheat germ agglutinin (an import inhibitor) and leptomycin B (an export inhibitor) induced premature senescence. These results suggest that Sp1 is an anti-senescence transcription factor in the telomere uncapping-induced senescence and that down-regulation of Sp1 leads to the senescence via down-regulation of the nuclear transport.
Collapse
|
10
|
Zeng X, Xu Z, Gu J, Huang H, Gao G, Zhang X, Li J, Jin H, Jiang G, Sun H, Huang C. Induction of miR-137 by Isorhapontigenin (ISO) Directly Targets Sp1 Protein Translation and Mediates Its Anticancer Activity Both In Vitro and In Vivo. Mol Cancer Ther 2016; 15:512-22. [PMID: 26832795 DOI: 10.1158/1535-7163.mct-15-0606] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/30/2015] [Indexed: 01/30/2023]
Abstract
Our recent studies found that isorhapontigenin (ISO) showed a significant inhibitory effect on human bladder cancer cell growth, accompanied with cell-cycle G0-G1 arrest as well as downregulation of Cyclin D1 expression at transcriptional level via inhibition of Sp1 transactivation in bladder cancer cells. In the current study, the potential ISO inhibition of bladder tumor formation has been explored in a xenograft nude mouse model, and the molecular mechanisms underlying ISO inhibition of Sp1 expression and anticancer activities have been elucidated both in vitro and in vivo. Moreover, the studies demonstrated that ISO treatment induced the expression of miR-137, which in turn suppressed Sp1 protein translation by directly targeting Sp1 mRNA 3'-untranslated region (UTR). Similar to ISO treatment, ectopic expression of miR-137 alone led to G0-G1 cell growth arrest and inhibition of anchorage-independent growth in human bladder cancer cells, which could be completely reversed by overexpression of GFP-Sp1. The inhibition of miR-137 expression attenuated ISO-induced inhibition of Sp1/Cyclin D1 expression, induction of G0-G1 cell growth arrest, and suppression of cell anchorage-independent growth. Taken together, our studies have demonstrated that miR-137 induction by ISO targets Sp1 mRNA 3'-UTR and inhibits Sp1 protein translation, which consequently results in reduction of Cyclin D1 expression, induction of G0-G1 growth arrest, and inhibition of anchorage-independent growth in vitro and in vivo. Our results have provided novel insights into understanding the anticancer activity of ISO in the therapy of human bladder cancer.
Collapse
Affiliation(s)
- Xingruo Zeng
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York. Department of Nephrology, Central Hospital of Wuhan, Wuhan, China
| | - Zhou Xu
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Jiayan Gu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haishan Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York. Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guangxun Gao
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Xiaoru Zhang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Honglei Jin
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Guosong Jiang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Hong Sun
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo, New York.
| |
Collapse
|
11
|
Li X, Pathi SS, Safe S. Sulindac sulfide inhibits colon cancer cell growth and downregulates specificity protein transcription factors. BMC Cancer 2015; 15:974. [PMID: 26673922 PMCID: PMC4682223 DOI: 10.1186/s12885-015-1956-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Specificity protein (Sp) transcription factors play pivotal roles in maintaining the phenotypes of many cancers. We hypothesized that the antineoplastic effects of sulindac and its metabolites were due, in part, to targeting downregulation of Sp transcription factors. METHODS The functional effects of sulindac, sulindac sulfone and sulindac sulfide on colon cancer cell proliferation were determined by cell counting. Effects of these compounds on expression of Sp1, Sp3, Sp4 and pro-oncogenic Sp-regulated genes were determined by western blot analysis of whole cell lysates and in transient transfection assays using GC-rich constructs. RESULTS Sulindac and its metabolites inhibited RKO and SW480 colon cancer cell growth and the order of growth inhibitory potency was sulindac sulfide>>sulindac sulfone>sulindac. Treatment of SW480 and RKO cells with sulindac sulfide downregulated expression of Sp1, Sp3 and Sp4 proteins. Sulindac sulfide also decreased expression of several Sp-regulated genes that are critical for cancer cell survival, proliferation and angiogenesis and these include survivin, bcl-2, epidermal growth factor receptor (EGFR), cyclin D1, p65 subunit of NFκB and vascular endothelial growth factor (VEGF). Sulindac sulfide also induced reactive oxygen species (ROS) and decreased the level of microRNA-27a in colon cancer cells, which resulted in the upregulation of the Sp-repressor ZBTB10 and this resulted in downregulation of Sp proteins. CONCLUSIONS The results suggest that the cancer chemotherapeutic effects of sulindac in colon cancer cells are due, in part, to its metabolite sulindac sulfide which downregulates Sp transcription factors and Sp-regulated pro-oncogenic gene products.
Collapse
Affiliation(s)
- Xi Li
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA
| | - Satya S Pathi
- Oklahoma Medical Research Foundation, 825 NE 13th St., Oklahoma City, OK, 73104, USA
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX, 77843-4466, USA.
| |
Collapse
|
12
|
Hedrick E, Crose L, Linardic CM, Safe S. Histone Deacetylase Inhibitors Inhibit Rhabdomyosarcoma by Reactive Oxygen Species-Dependent Targeting of Specificity Protein Transcription Factors. Mol Cancer Ther 2015; 14:2143-2153. [PMID: 26162688 PMCID: PMC4618474 DOI: 10.1158/1535-7163.mct-15-0148] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/25/2015] [Indexed: 11/16/2022]
Abstract
The two major types of rhabdomyosarcoma (RMS) are predominantly diagnosed in children, namely embryonal (ERMS) and alveolar (ARMS) RMS, and patients are treated with cytotoxic drugs, which results in multiple toxic side effects later in life. Therefore, development of innovative chemotherapeutic strategies is imperative, and a recent genomic analysis suggested the potential efficacy of reactive oxygen species (ROS)-inducing agents. Here, we demonstrate the efficacy of the potent histone deacetylase (HDAC) inhibitors, panobinostat and vorinostat, as agents that inhibit RMS tumor growth in vivo, induce apoptosis, and inhibit invasion of RD and Rh30 RMS cell lines. These effects are due to epigenetic repression of cMyc, which leads to decreased expression of cMyc-regulated miRs-17, -20a, and -27a; upregulation of ZBTB4, ZBTB10, and ZBTB34; and subsequent downregulation of Sp transcription factors. We also show that inhibition of RMS cell growth, survival and invasion, and repression of Sp transcription factors by the HDAC inhibitors are independent of histone acetylation but reversible after cotreatment with the antioxidant glutathione. These results show a novel ROS-dependent mechanism of antineoplastic activity for panobinostat and vorinostat that lies outside of their canonical HDAC-inhibitory activity and demonstrates the potential clinical utility for treating RMS patients with ROS-inducing agents.
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Lisa Crose
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina
| | - Corinne M Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina. Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas. Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, Texas.
| |
Collapse
|
13
|
Safe S. MicroRNA-Specificity Protein (Sp) Transcription Factor Interactions and Significance in Carcinogenesis. CURRENT PHARMACOLOGY REPORTS 2015; 1:73-78. [PMID: 26457240 PMCID: PMC4596546 DOI: 10.1007/s40495-014-0012-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Specificity protein (Sp) transcription factors (TFs) such as Sp1, Sp3 and Sp4 are overexpressed in tumors and Sp1 is a negative prognostic factor for multiple tumor types. Sp TFs regulate expression of pro-oncogenic factors important for cell proliferation, survival, angiogenesis, migration/invasion and inflammation and the high expression of Sp TFs in tumors is primarily due to miRNAs. For example, expression of tumor-suppressor-like miRNAs such as miR-200b/c, miR-335, miR-22, miR-149 and others that inactivate Sp1 expression is low in many tumor types. Research in our laboratory has also demonstrated that high expression of Sp TFs is also due to miRNA-dependent inhibition of the transcriptional repressors ZBTB10 and ZBTB4 by miR-27a and miR-20a/miR-17p, respectively. Thus, miRNAs play a critical role in maintaining high levels of Sp1, Sp3, Sp4 and pro-oncogenic Sp-regulated genes in tumors and cancer cells, and there is ample evidence that anticancer agents targeting the miRNASp TF axis can be highly effective for cancer chemotherapy.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466 USA, Tel: 979-845-5989, ,
| |
Collapse
|
14
|
Safe S, Imanirad P, Sreevalsan S, Nair V, Jutooru I. Transcription factor Sp1, also known as specificity protein 1 as a therapeutic target. Expert Opin Ther Targets 2014; 18:759-769. [PMID: 24793594 DOI: 10.1517/14728222.2014.914173] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Specificity protein (Sp) transcription factors (TFs) are members of the Sp/Kruppel-like factor family, and Sp proteins play an important role in embryonic and early postnatal development. Sp1 has been the most extensively investigated member of this family, and expression of this protein decreases with age, whereas Sp1 and other family members (Sp3 and Sp4) are highly expressed in tumors and cancer cell lines. AREA COVERED The prognostic significance of Sp1 in cancer patients and the functional pro-oncogenic activities of Sp1, Sp3 and Sp4 in cancer cell lines are summarized. Several different approaches have been used to target downregulation of Sp TFs and Sp-regulated genes, and this includes identification of different structural classes of antineoplastic agents including NSAIDs, natural products and their synthetic analogs and several well-characterized drugs including arsenic trioxide, aspirin and metformin. The multiple pathways involved in drug-induced Sp downregulation are also discussed. EXPERT OPINION The recognition by the scientific and clinical community that experimental and clinically used antineoplastic agents downregulate Sp1, Sp3 and Sp4, and pro-oncogenic Sp-regulated genes will facilitate future clinical applications for individual drug and drug combination therapies that take advantage of their unusual effects.
Collapse
Affiliation(s)
- Stephen Safe
- Texas A&M University, Veterinary Physiology and Pharmacology , 4466 TAMU, College Station, TX 77843-4466 , USA
| | | | | | | | | |
Collapse
|
15
|
Pathi S, Li X, Safe S. Tolfenamic acid inhibits colon cancer cell and tumor growth and induces degradation of specificity protein (Sp) transcription factors. Mol Carcinog 2014; 53 Suppl 1:E53-E61. [PMID: 23670891 DOI: 10.1002/mc.22010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/20/2012] [Accepted: 01/04/2013] [Indexed: 11/09/2022]
Abstract
Tolfenamic acid (TA) is a non-steroidal anti-inflammatory drug (NSAID) that inhibits lung, esophageal, breast and pancreatic cancer cell and tumor growth, and this study investigated the anticancer activity of TA in colon cancer. TA inhibited growth and induced apoptosis in RKO, SW480, HT-29, and HCT-116 colon cancer cells, and TA (50 mg/kg/d) also inhibited tumor growth in athymic nude mice bearing RKO cells as xenografts. TA downregulated expression of Sp proteins (Sp1, Sp3, and Sp4) in colon cancer cells and this was accompanied by decreased expression of several Sp-regulated growth promoting (cyclin D1, hepatocyte growth factor receptor), angiogenic (vascular endothelial growth factor (VEGF) and its receptor 1), survival (survivin and bcl-2), and inflammatory (NFκBp65/p50) gene products. The mechanism of TA-mediated effects on Sp proteins was due to activation of caspases. These results now extend the number of NSAIDs that may have clinical potential for colon cancer chemotherapy and show that the anticancer activity of TA is due, in part, to targeting Sp transcription factors.
Collapse
Affiliation(s)
- Satya Pathi
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | | | | |
Collapse
|
16
|
Abstract
Several agents used for treatment of colon and other cancers induce reactive oxygen species (ROS) and this plays an important role in their anticancer activities. In addition to the well-known proapoptotic effects of ROS inducers, these compounds also decrease expression of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and several pro-oncogenic Spregulated genes important for cancer cell proliferation, survival and metastasis. The mechanism of these responses involve ROS-dependent downregulation of microRNA-27a (miR-27a) or miR-20a (and paralogs) and induction of two Sp-repressors, ZBTB10 and ZBTB4 respectively. This pathway significantly contributes to the anticancer activity of ROS inducers and should be considered in development of drug combinations for cancer chemotherapy.
Collapse
Affiliation(s)
- Sandeep Sreevalsan
- VMR 1197, Room 413, Texas A&M University, College Station, TX, 77843 979-845-9182
| | - Stephen Safe
- VMR 1197, Room 410, Texas A&M University, College Station, TX, 77843 979-845-5988
| |
Collapse
|
17
|
Mertens-Talcott SU, Noratto GD, Li X, Angel-Morales G, Bertoldi MC, Safe S. Betulinic acid decreases ER-negative breast cancer cell growth in vitro and in vivo: role of Sp transcription factors and microRNA-27a:ZBTB10. Mol Carcinog 2013; 52:591-602. [PMID: 22407812 PMCID: PMC3418350 DOI: 10.1002/mc.21893] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 01/27/2012] [Accepted: 02/06/2012] [Indexed: 11/10/2022]
Abstract
Betulinic acid (BA), a pentacyclic triterpenoid isolated from tree bark is cytotoxic to cancer cells. There is evidence that specificity proteins (Sps), such as Sp1, Sp3, and Sp4, are overexpressed in tumors and contribute to the proliferative and angiogenic phenotype associated with cancer cells. The objective of this study was to determine the efficacy of BA in decreasing the Sps expression and underlying mechanisms. Results show that BA decreased proliferation and induced apoptosis of estrogen-receptor-negative breast cancer MDA-MB-231 cells. The BA-induced Sp1, Sp3, and Sp4 downregulation was accompanied by increased zinc finger ZBTB10 expression, a putative Sp-repressor and decreased microRNA-27a levels, a microRNA involved in the regulation of ZBTB10. Similar results were observed in MDA-MB-231 cells transfected with ZBTB10 expression plasmid. BA induced cell cycle arrest in the G2/M phase and increased Myt-1 mRNA (a microRNA-27a target gene), which causes inhibition in G2/M by phosphorylation of cdc2. The effects of BA were reversed by transient transfection with a mimic of microRNA-27a. In nude mice with xenografted MDA-MB-231 cells, tumor size and weight were significantly decreased by BA treatment. In tumor tissue, ZBTB10 mRNA was increased while mRNA and protein of Sp1, Sp3 and Sp4, as well as mRNA of vascular endothelial growth factor receptor (VEGFR), survivin and microRNA-27a were decreased by BA. In lungs of xenografted mice, human β2-microglobulin mRNA was decreased in BA-treated animals. These results show that the anticancer effects of BA are at least in part based on interactions with the microRNA-27a-ZBTB10-Sp-axis causing increased cell death.
Collapse
Affiliation(s)
- Susanne U. Mertens-Talcott
- Department of Nutrition & Food Science
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine
- Institute for Obesity Research and Program Evaluation, Texas A&M University College Station, TX 77843
| | - Giuliana D. Noratto
- Department of Nutrition & Food Science
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine
- Institute for Obesity Research and Program Evaluation, Texas A&M University College Station, TX 77843
| | | | | | | | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine
- Center for Environmental and Genetic Medicine Texas A&M Health Science Center Institute of Biosciences and Technology Houston, TX
| |
Collapse
|
18
|
Chadalapaka G, Jutooru I, Sreevalsan S, Pathi S, Kim K, Chen C, Crose L, Linardic C, Safe S. Inhibition of rhabdomyosarcoma cell and tumor growth by targeting specificity protein (Sp) transcription factors. Int J Cancer 2013; 132:795-806. [PMID: 22815231 PMCID: PMC3527649 DOI: 10.1002/ijc.27730] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 06/08/2012] [Indexed: 12/30/2022]
Abstract
Specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 are highly expressed in rhabdomyosarcoma (RMS) cells. In tissue arrays of RMS tumor cores from 71 patients, 80% of RMS patients expressed high levels of Sp1 protein, whereas low expression of Sp1 was detected in normal muscle tissue. The non-steroidal anti-inflammatory drug (NSAID) tolfenamic acid (TA) inhibited growth and migration of RD and RH30 RMS cell lines and also inhibited tumor growth in vivo using a mouse xenograft (RH30 cells) model. The effects of TA were accompanied by downregulation of Sp1, Sp3, Sp4 and Sp-regulated genes in RMS cells and tumors, and the role of Sp protein downregulation in mediating inhibition of RD and RH30 cell growth and migration was confirmed by individual and combined knockdown of Sp1, Sp3 and Sp4 proteins by RNA interference. TA treatment and Sp knockdown in RD and RH30 cells also showed that four genes that are emerging as individual drug targets for treating RMS, namely c-MET, insulin-like growth factor receptor (IGFR), PDGFRα and CXCR4, are also Sp-regulated genes. These results suggest that NSAIDs such as TA may have potential clinical efficacy in drug combinations for treating RMS patients.
Collapse
Affiliation(s)
- Gayathri Chadalapaka
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
| | - Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
| | - Sandeep Sreevalsan
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
| | - Satya Pathi
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
| | - Kyounghyun Kim
- Institute for Biosciences and Technology, Texas A&M Health Science Center, Houston, TX
| | - Candy Chen
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Lisa Crose
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Corinne Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
- Institute for Biosciences and Technology, Texas A&M Health Science Center, Houston, TX
| |
Collapse
|
19
|
Kim SY, Kang HT, Han JA, Park SC. The transcription factor Sp1 is responsible for aging-dependent altered nucleocytoplasmic trafficking. Aging Cell 2012; 11:1102-9. [PMID: 23013401 DOI: 10.1111/acel.12012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2012] [Indexed: 01/23/2023] Open
Abstract
Hyporesponsiveness to external signals, such as growth factors and apoptotic stimuli, is a cardinal feature of cellular senescence. We previously reported that an aging-dependent marked reduction in nucleocytoplasmic trafficking (NCT)-related genes could be responsible for this phenomenon. In searching for the mechanism, we identified the transcription factor, Sp1, as a common regulator of NCT genes, including various nucleoporins, importins, exportins, and Ran GTPase cycle-related genes. Sp1 knockdown led to a reduction of those genes in young human diploid fibroblast cells (HDF); Sp1 overexpression induced those genes in senescent cells. In addition, epidermal growth factor stimulation-induced p-ERK1/2 nuclear translocation and Elk-1 phosphorylation were severely impaired by Sp1 depletion in young HDFs; Sp1 overexpression restored the nuclear translocation of p-ERK1/2 in senescent HDFs. Furthermore, we observed that Sp1 protein levels were decreased in senescent cells, and H(2) O(2) treatment decreased Sp1 levels in a proteasome-dependent manner. In addition, O-GlcNAcylation of Sp1 was decreased in senescent cells as well as in H(2) O(2) -treated cells. Taken together, these results suggest that Sp1 could be a key regulator in the control of NCT genes and that reactive oxygen species-mediated alteration in Sp1 stability may be responsible for the generalized repression of those genes, leading to formation of the senescence-dependent functional nuclear barrier, resulting in subsequent hyporesponsiveness to external signals.
Collapse
Affiliation(s)
| | - Hyun T. Kang
- Lee Gil Ya Cancer and Diabetes Institute; Gachon University; Incheon; 406-840; South Korea
| | - Jeong A. Han
- Department of Biochemistry and Molecular Biology; School of Medicine; Kangwon National University; Chuncheon; 200-701; South Korea
| | | |
Collapse
|
20
|
Gandhy SU, Kim K, Larsen L, Rosengren RJ, Safe S. Curcumin and synthetic analogs induce reactive oxygen species and decreases specificity protein (Sp) transcription factors by targeting microRNAs. BMC Cancer 2012; 12:564. [PMID: 23194063 PMCID: PMC3522018 DOI: 10.1186/1471-2407-12-564] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 11/23/2012] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Curcumin inhibits growth of several cancer cell lines, and studies in this laboratory in bladder and pancreatic cancer cells show that curcumin downregulates specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and pro-oncogenic Sp-regulated genes. In this study, we investigated the anticancer activity of curcumin and several synthetic cyclohexanone and piperidine analogs in colon cancer cells. METHODS The effects of curcumin and synthetic analogs on colon cancer cell proliferation and apoptosis were determined using standardized assays. The changes in Sp proteins and Sp-regulated gene products were analysed by western blots, and real time PCR was used to determine microRNA-27a (miR-27a), miR-20a, miR-17-5p and ZBTB10 and ZBTB4 mRNA expression. RESULTS The IC50 (half-maximal) values for growth inhibition (24 hr) of colon cancer cells by curcumin and synthetic cyclohexanone and piperidine analogs of curcumin varied from 10 μM for curcumin to 0.7 μM for the most active synthetic piperidine analog RL197, which was used along with curcumin as model agents in this study. Curcumin and RL197 inhibited RKO and SW480 colon cancer cell growth and induced apoptosis, and this was accompanied by downregulation of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and Sp-regulated genes including the epidermal growth factor receptor (EGFR), hepatocyte growth factor receptor (c-MET), survivin, bcl-2, cyclin D1 and NFκB (p65 and p50). Curcumin and RL197 also induced reactive oxygen species (ROS), and cotreatment with the antioxidant glutathione significantly attenuated curcumin- and RL197-induced growth inhibition and downregulation of Sp1, Sp3, Sp4 and Sp-regulated genes. The mechanism of curcumin-/RL197-induced repression of Sp transcription factors was ROS-dependent and due to induction of the Sp repressors ZBTB10 and ZBTB4 and downregulation of microRNAs (miR)-27a, miR-20a and miR-17-5p that regulate these repressors. CONCLUSIONS These results identify a new and highly potent curcumin derivative and demonstrate that in cells where curcumin and RL197 induce ROS, an important underlying mechanism of action involves perturbation of miR-ZBTB10/ZBTB4, resulting in the induction of these repressors which downregulate Sp transcription factors and Sp-regulated genes.
Collapse
Affiliation(s)
- Shruti U Gandhy
- College of Medicine, Texas A&M Health Sciences Center, Houston, TX, 77030, USA
| | - KyoungHyun Kim
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd, Houston, TX, 77030, USA
| | - Lesley Larsen
- Department of Chemistry, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, P.O. Box 913, Dunedin, 9054, New Zealand
| | - Stephen Safe
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd, Houston, TX, 77030, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| |
Collapse
|
21
|
Kim K, Chadalapaka G, Pathi SS, Jin UH, Lee JS, Park YY, Cho SG, Chintharlapalli S, Safe S. Induction of the transcriptional repressor ZBTB4 in prostate cancer cells by drug-induced targeting of microRNA-17-92/106b-25 clusters. Mol Cancer Ther 2012; 11:1852-1862. [PMID: 22752225 PMCID: PMC3632183 DOI: 10.1158/1535-7163.mct-12-0181] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Androgen-insensitive DU145 and PC3 human prostate cancer cells express high levels of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and treatment of cells with methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate (CDODA-Me) inhibited cell growth and downregulated Sp1, Sp3, and Sp4 expression. CDODA-Me (15 mg/kg/d) was a potent inhibitor of tumor growth in a mouse xenograft model (PC3 cells) and also decreased expression of Sp transcription factors in tumors. CDODA-Me-mediated downregulation of Sp1, Sp3, and Sp4 was due to induction of the transcriptional repressor ZBTB4, which competitively binds and displaces Sp transcription factors from GC-rich sites in Sp1-, Sp3-, Sp4-, and Sp-regulated gene promoters. ZBTB4 levels are relatively low in DU145 and PC3 cells due to suppression by miR paralogs that are members of the miR-17-92 (miR-20a/17-5p) and miR-106b-25 (miR-106b/93) clusters. Examination of publically available prostate cancer patient array data showed an inverse relationship between ZBTB4 and miRs-20a/17-5p/106b/93 expression, and increased ZBTB4 in patients with prostate cancer was a prognostic factor for increased survival. CDODA-Me induces ZBTB4 in prostate cancer cells through disruption of miR-ZBTB4 interactions, and this results in downregulation of pro-oncogenic Sp transcription factors and Sp-regulated genes.
Collapse
Affiliation(s)
- KyoungHyun Kim
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX, 77030
| | - Gayathri Chadalapaka
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843
| | - Satya S. Pathi
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843
| | - Un-Ho Jin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX, 77030
| | - Ju-Seog Lee
- Department of Systems Biology, M.D. Anderson Cancer Center, The University of Texas, 1515 Holcombe Blvd., Unit 950, Houston, TX 77030
| | - Yun-Yong Park
- Department of Systems Biology, M.D. Anderson Cancer Center, The University of Texas, 1515 Holcombe Blvd., Unit 950, Houston, TX 77030
| | - Sung-Gook Cho
- Department of Systems Biology, M.D. Anderson Cancer Center, The University of Texas, 1515 Holcombe Blvd., Unit 950, Houston, TX 77030
| | - Sudhakar Chintharlapalli
- Eli Lilly and Company, Lilly Research Labs - Oncology Division, DC0546 Room H48A-4105, Indianapolis, IN 46285
| | - Stephen Safe
- Institute of Biosciences and Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Houston, TX, 77030
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843
| |
Collapse
|
22
|
Pathi SS, Lei P, Sreevalsan S, Chadalapaka G, Jutooru I, Safe S. Pharmacologic doses of ascorbic acid repress specificity protein (Sp) transcription factors and Sp-regulated genes in colon cancer cells. Nutr Cancer 2011; 63:1133-1142. [PMID: 21919647 PMCID: PMC3359146 DOI: 10.1080/01635581.2011.605984] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ascorbic acid (vitamin C) inhibits cancer cell growth, and there is a controversy regarding the cancer chemoprotective effects of pharmacologic doses of this compound that exhibits prooxidant activity. We hypothesized that the anticancer activity of pharmacologic doses of ascorbic acid (<5 mM) is due, in part, to reactive oxygen species-dependent downregulation of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 and Sp-regulated genes. In this study, ascorbic acid (1-3 mM) decreased RKO and SW480 colon cancer cell proliferation and induced apoptosis and necrosis, and this was accompanied by downregulation of Sp1, Sp3, and Sp4 proteins. In addition, ascorbic acid decreased expression of several Sp-regulated genes that are involved in cancer cell proliferation [hepatocyte growth factor receptor (c-Met), epidermal growth factor receptor and cyclin D1], survival (survivin and bcl-2), and angiogenesis [vascular endothelial growth factor (VEGF) and its receptors (VEGFR1 and VEGFR2)]. Other prooxidants such as hydrogen peroxide exhibited similar activities in colon cancer cells, and cotreatment with glutathione inhibited these responses. This study demonstrates for the first time that the anticancer activities of ascorbic acid are due, in part, to ROS-dependent repression of Sp transcription factors.
Collapse
Affiliation(s)
- Satya S. Pathi
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Ping Lei
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030
| | - Sandeep Sreevalsan
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Gayathri Chadalapaka
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Indira Jutooru
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843-4466
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030
| |
Collapse
|
23
|
Chintharlapalli S, Papineni S, Lei P, Pathi S, Safe S. Betulinic acid inhibits colon cancer cell and tumor growth and induces proteasome-dependent and -independent downregulation of specificity proteins (Sp) transcription factors. BMC Cancer 2011; 11:371. [PMID: 21864401 PMCID: PMC3170653 DOI: 10.1186/1471-2407-11-371] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/24/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Betulinic acid (BA) inhibits growth of several cancer cell lines and tumors and the effects of BA have been attributed to its mitochondriotoxicity and inhibition of multiple pro-oncogenic factors. Previous studies show that BA induces proteasome-dependent degradation of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 in prostate cancer cells and this study focused on the mechanism of action of BA in colon cancer cells. METHODS The effects of BA on colon cancer cell proliferation and apoptosis and tumor growth in vivo were determined using standardized assays. The effects of BA on Sp proteins and Sp-regulated gene products were analyzed by western blots, and real time PCR was used to determine microRNA-27a (miR-27a) and ZBTB10 mRNA expression. RESULTS BA inhibited growth and induced apoptosis in RKO and SW480 colon cancer cells and inhibited tumor growth in athymic nude mice bearing RKO cells as xenograft. BA also decreased expression of Sp1, Sp3 and Sp4 transcription factors which are overexpressed in colon cancer cells and decreased levels of several Sp-regulated genes including survivin, vascular endothelial growth factor, p65 sub-unit of NFκB, epidermal growth factor receptor, cyclin D1, and pituitary tumor transforming gene-1. The mechanism of action of BA was dependent on cell context, since BA induced proteasome-dependent and proteasome-independent downregulation of Sp1, Sp3 and Sp4 in SW480 and RKO cells, respectively. In RKO cells, the mechanism of BA-induced repression of Sp1, Sp3 and Sp4 was due to induction of reactive oxygen species (ROS), ROS-mediated repression of microRNA-27a, and induction of the Sp repressor gene ZBTB10. CONCLUSIONS These results suggest that the anticancer activity of BA in colon cancer cells is due, in part, to downregulation of Sp1, Sp3 and Sp4 transcription factors; however, the mechanism of this response is cell context-dependent.
Collapse
Affiliation(s)
- Sudhakar Chintharlapalli
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030 USA
- Eli Lilly Co., Oncology Division, Indianapolis, IN, USA
| | - Sabitha Papineni
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030 USA
- Dow Agrosciences, Indianapolis, IN, USA
| | - Ping Lei
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030 USA
| | - Satya Pathi
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843 USA
| | - Stephen Safe
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
24
|
Li S, Lu L, Hao S, Wang Y, Zhang L, Liu S, Liu B, Li K, Luo X. Dietary manganese modulates expression of the manganese-containing superoxide dismutase gene in chickens. J Nutr 2011; 141:189-94. [PMID: 21169227 DOI: 10.3945/jn.110.126680] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To investigate the possible mechanism(s) by which dietary manganese (Mn) levels and sources modulate the expression of the manganese-containing superoxide dismutase (MnSOD) gene at both the transcriptional and translational levels, we used 432 8-d-old male broiler chicks in a 1 plus 4 × 2 design. Chickens were given either a diet without Mn supplementation [control (C)] or diets supplemented with 100 (optimal) or 200 (high) mg Mn/kg diet from inorganic Mn sulfate (I) or 3 organic complexes of Mn and amino acids with weak (W), moderate (M), or strong (S) chelation strength up to 21 d of age. Compared with C chicks, chicks fed Mn-supplemented diets had higher (P < 0.01) Mn concentrations, specificity protein 1 (Sp1) DNA-binding activities, MnSOD mRNA levels, MnSOD mRNA-binding protein (MnSOD-BP) RNA-binding activities, MnSOD protein concentrations, and MnSOD activities within heart tissue, but lower (P < 0.01) heart activating protein-2 (AP-2) DNA-binding activities. Chicks fed M diets had higher (P < 0.05) heart Mn concentrations, MnSOD mRNA levels, and MnSOD-BP RNA-binding activities compared with those fed the I and W diets and lower (P < 0.01) AP-2 DNA-binding activities than those fed other treatment diets. These results suggest that dietary Mn could modulate the expression of the MnSOD gene in broilers by altering Sp1 and AP-2 DNA-binding activities at the transcriptional level and enhancing MnSOD-BP RNA-binding activity at the translational level. Additionally, an organic Mn source with moderate chelation strength could be more effective than other Mn sources in activating MnSOD gene expression at both the transcriptional and translational levels.
Collapse
Affiliation(s)
- Sufen Li
- Mineral Nutrition Research Division, State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, P R China
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
Jutooru I, Chadalapaka G, Abdelrahim M, Basha MR, Samudio I, Konopleva M, Andreeff M, Safe S. Methyl 2-cyano-3,12-dioxooleana-1,9-dien-28-oate decreases specificity protein transcription factors and inhibits pancreatic tumor growth: role of microRNA-27a. Mol Pharmacol 2010; 78:226-236. [PMID: 20488920 PMCID: PMC2917860 DOI: 10.1124/mol.110.064451] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 05/13/2010] [Indexed: 01/11/2023] Open
Abstract
The anticancer agent 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and its methyl ester (CDDO-Me) typically induce a broad spectrum of growth-inhibitory, proapoptotic, and antiangiogenic responses. Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. Repression of Sp and Sp-dependent genes by CDDO-Me was due to the down-regulation of microRNA-27a and induction of zinc finger and BTB domain containing 10 (ZBTB10), an Sp repressor, and these responses were also reversed by antioxidants. Thus, the anticancer activity of CDDO-Me is due, in part, to activation of ROS, which in turn targets the microRNA-27a:ZBTB10-Sp transcription factor axis. This results in decreased expression of Sp-regulated genes, growth inhibition, induction of apoptosis, and antiangiogenic responses.
Collapse
Affiliation(s)
- Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, Vet. Res. Bldg. 410, College Station, TX 77843-4466, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jutooru I, Chadalapaka G, Sreevalsan S, Lei P, Barhoumi R, Burghardt R, Safe S. Arsenic trioxide downregulates specificity protein (Sp) transcription factors and inhibits bladder cancer cell and tumor growth. Exp Cell Res 2010; 316:2174-88. [PMID: 20435036 PMCID: PMC2900380 DOI: 10.1016/j.yexcr.2010.04.027] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 04/05/2010] [Accepted: 04/26/2010] [Indexed: 11/18/2022]
Abstract
Arsenic trioxide exhibits antiproliferative, antiangiogenic and proapoptotic activity in cancer cells, and many genes associated with these responses are regulated by specificity protein (Sp) transcription factors. Treatment of cancer cells derived from urologic (bladder and prostate) and gastrointestinal (pancreas and colon) tumors with arsenic trioxide demonstrated that these cells exhibited differential responsiveness to the antiproliferative effects of this agent and this paralleled their differential repression of Sp1, Sp3 and Sp4 proteins in the same cell lines. Using arsenic trioxide-responsive KU7 and non-responsive 253JB-V bladder cancer cells as models, we show that in KU7 cells, < or =5 microM arsenic trioxide decreased Sp1, Sp3 and Sp4 and several Sp-dependent genes and responses including cyclin D1, epidermal growth factor receptor, bcl-2, survivin and vascular endothelial growth factor, whereas at concentrations up to 15 microM, minimal effects were observed in 253JB-V cells. Arsenic trioxide also inhibited tumor growth in athymic mice bearing KU7 cells as xenografts, and expression of Sp1, Sp3 and Sp4 was significantly decreased. Inhibitors of oxidative stress such as glutathione or dithiothreitol protected KU7 cells from arsenic trioxide-induced antiproliferative activity and Sp repression, whereas glutathione depletion sensitized 253JB-V cells to arsenic trioxide. Mechanistic studies suggested that arsenic trioxide-dependent downregulation of Sp and Sp-dependent genes was due to decreased mitochondrial membrane potential and induction of reactive oxygen species, and the role of peroxides in mediating these responses was confirmed using hydrogen peroxide.
Collapse
Affiliation(s)
- Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA
| | - Gayathri Chadalapaka
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA
| | - Sandeep Sreevalsan
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA
| | - Ping Lei
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, TX 77030-3303 USA
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Robert Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466 USA
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, TX 77030-3303 USA
| |
Collapse
|
28
|
Paquet C, Larouche D, Bisson F, Proulx S, Simard-Bisson C, Gaudreault M, Robitaille H, Carrier P, Martel I, Duranceau L, Auger FA, Fradette J, Guérin SL, Germain L. Tissue engineering of skin and cornea: Development of new models for in vitro studies. Ann N Y Acad Sci 2010; 1197:166-77. [PMID: 20536846 DOI: 10.1111/j.1749-6632.2009.05373.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human beings are greatly preoccupied with the unavoidable nature of aging. While the biological processes of senescence and aging are the subjects of intense investigations, the molecular mechanisms linking aging with disease and death are yet to be elucidated. Tissue engineering offers new models to study the various processes associated with aging. Using keratin 19 as a stem cell marker, our studies have revealed that stem cells are preserved in human skin reconstructed by tissue engineering and that the number of epithelial stem cells varies according to the donor's age. As with skin, human corneas can also be engineered in vitro. Among the epithelial cells used for reconstructing skin and corneas, significant age-dependent variations in the expression of the transcription factor Sp1 were observed. Culturing skin epithelial cells with a feeder layer extended their life span in culture, likely by preventing Sp1 degradation in epithelial cells, therefore demonstrating the pivotal role played by this transcription factor in cell proliferation. Finally, using the human tissue-engineered skin as a model, we linked Hsp27 activation with skin differentiation.
Collapse
Affiliation(s)
- Claudie Paquet
- Laboratoire d'Organogénèse Expérimentale, Centre de Recherche FRSQ du CHA Universitaire de Québec, and Département de Chirurgie et d'Oto-rhino-laryngologie et Ophtalmologie, Université Laval, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chadalapaka G, Jutooru I, Burghardt R, Safe S. Drugs that target specificity proteins downregulate epidermal growth factor receptor in bladder cancer cells. Mol Cancer Res 2010; 8:739-50. [PMID: 20407012 PMCID: PMC2872686 DOI: 10.1158/1541-7786.mcr-09-0493] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The epidermal growth factor receptor (EGFR) is an important chemotherapeutic target for tyrosine kinase inhibitors and antibodies that block the extracellular domain of EGFR. Betulinic acid (BA) and curcumin inhibited bladder cancer cell growth and downregulated specificity protein (Sp) transcription factors, and this was accompanied by decreased expression of EGFR mRNA and protein levels. EGFR, a putative Sp-regulated gene, was also decreased in cells transfected with a cocktail (iSp) containing small inhibitory RNAs for Sp1, Sp3, and Sp4, and RNA interference with individual Sp knockdown indicated that EGFR expression was primarily regulated by Sp1 and Sp3. BA, curcumin, and iSp also decreased phosphorylation of Akt in these cells, and downregulation of EGFR by BA, curcumin, and iSp was accompanied by induction of LC3 and autophagy, which is consistent with recent studies showing that EGFR suppresses autophagic cell death. The results show that EGFR is an Sp-regulated gene in bladder cancer, and drugs such as BA and curcumin that repress Sp proteins also ablate EGFR expression. Thus, compounds such as curcumin and BA that downregulate Sp transcription factors represent a novel class of anticancer drugs that target EGFR in bladder cancer cells and tumors by inhibiting receptor expression.
Collapse
Affiliation(s)
- Gayathri Chadalapaka
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
| | - Indira Jutooru
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
| | - Robert Burghardt
- Department of Veterinary Integrated Biosciences, Texas A&M University, College Station, TX 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030
| |
Collapse
|
30
|
Lukosz M, Jakob S, Büchner N, Zschauer TC, Altschmied J, Haendeler J. Nuclear redox signaling. Antioxid Redox Signal 2010; 12:713-42. [PMID: 19737086 DOI: 10.1089/ars.2009.2609] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species have been described to modulate proteins within the cell, a process called redox regulation. However, the importance of compartment-specific redox regulation has been neglected for a long time. In the early 1980s and 1990s, many in vitro studies introduced the possibility that nuclear redox signaling exists. However, the functional relevance for that has been greatly disregarded. Recently, it has become evident that nuclear redox signaling is indeed one important signaling mechanism regulating a variety of cellular functions. Transcription factors, and even kinases and phosphatases, have been described to be redox regulated in the nucleus. This review describes several of these proteins in closer detail and explains their functions resulting from nuclear localization and redox regulation. Moreover, the redox state of the nucleus and several important nuclear redox regulators [Thioredoxin-1 (Trx-1), Glutaredoxins (Grxs), Peroxiredoxins (Prxs), and APEX nuclease (multifunctional DNA-repair enzyme) 1 (APEX1)] are introduced more precisely, and their necessity for regulation of transcription factors is emphasized.
Collapse
Affiliation(s)
- Margarete Lukosz
- Molecular Cell & Aging Research, IUF (Institute for Molecular Preventive Medicine), At the University of Duesseldorf gGmbH, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Li H, Costantini C, Scrable H, Weindruch R, Puglielli L. Egr-1 and Hipk2 are required for the TrkA to p75(NTR) switch that occurs downstream of IGF1-R. Neurobiol Aging 2009; 30:2010-20. [PMID: 18378044 PMCID: PMC2776679 DOI: 10.1016/j.neurobiolaging.2008.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 02/14/2008] [Accepted: 02/14/2008] [Indexed: 02/07/2023]
Abstract
The aging program mediated by IGF1-R is responsible for a naturally occurring TrkA to p75(NTR) switch that leads to activation of the second messenger ceramide and increased production of the Alzheimer's disease amyloid beta-peptide. Biochemical and genetic approaches that target IGF1-R signaling, p75(NTR), or ceramide are able to block the above events. Here, we show that the transcription factors Egr-1 and Hipk2 are required elements for the TrkA to p75(NTR) switch downstream of IGF1-R signaling. Specifically, Egr-1 is required for the upregulation of p75(NTR), whereas Hipk2 is required for the downregulation of TrkA. In fact, gene silencing of Egr-1 abolished the ability of IGF1 to upregulate p75(NTR), whereas similar approaches directed against Hipk2 blocked the downregulation of TrkA. In addition, IGF1 treatment favored binding of Egr-1 and Hipk2 to the promoter of p75(NTR) and TrkA, respectively. Finally, the expression levels of both Egr-1 and Hipk2 are upregulated in an age-dependent fashion. Such an event is opposed by caloric restriction, a model of delayed aging, and favored by the p44 transgene in p44(+/+) animals, a model of accelerated aging.
Collapse
Affiliation(s)
- Hui Li
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Claudio Costantini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Heidi Scrable
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard Weindruch
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Geriatric Research Education Clinical Center, VA Medical Center, Madison, Wisconsin 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- Geriatric Research Education Clinical Center, VA Medical Center, Madison, Wisconsin 53705, USA
| |
Collapse
|
32
|
Landriscina M, Maddalena F, Laudiero G, Esposito F. Adaptation to oxidative stress, chemoresistance, and cell survival. Antioxid Redox Signal 2009; 11:2701-16. [PMID: 19778285 DOI: 10.1089/ars.2009.2692] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The discovery of some additional properties and functions of reactive oxygen species (ROS), beyond their toxic effects, provides a novel scenario for the molecular basis and cell regulation of several pathophysiologic processes. ROS are generated by redox-sensitive, prosurvival signaling pathways and function as second messengers in the transduction of several extracellular signals. A complex intracellular redox buffering network has developed to adapt and protect cells against the dangerous effects of oxidative stress. However, pathways involved in ROS-adaptive response may also play a critical role in protecting cells against cytotoxic effects of anticancer agents, thus supporting the hypothesis of a correlation between adaptation/resistance to oxidative stress and resistance to anticancer drugs. This review summarizes the main systems involved in the adaptive responses: an overview on the pathophysiologic relevance of mitochondria on redox-sensitive transcription factors and genes and main antioxidant networks in tumor cells is provided. One of the major aims is to highlight the adaptive mechanisms and their interplay in the intricate connection between oncogenic signaling, oxidative stress, and chemoresistance. Clarification of these mechanisms has tremendous application potential, in terms of developing novel molecular-targeted anticancer therapies and innovative strategies for rational combination of these agents with chemotherapeutic or tumor-specific biologic drugs.
Collapse
Affiliation(s)
- Matteo Landriscina
- Clinical Oncology Unit, Department of Medical Sciences, University of Foggia, Foggia, Italy
| | | | | | | |
Collapse
|
33
|
Papineni S, Chintharlapalli S, Abdelrahim M, Lee SO, Burghardt R, Abudayyeh A, Baker C, Herrera L, Safe S. Tolfenamic acid inhibits esophageal cancer through repression of specificity proteins and c-Met. Carcinogenesis 2009; 30:1193-1201. [PMID: 19406933 PMCID: PMC2704282 DOI: 10.1093/carcin/bgp092] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 04/06/2009] [Accepted: 04/09/2009] [Indexed: 12/15/2022] Open
Abstract
The non-steroidal anti-inflammatory drug tolfenamic acid (TA) inhibits proliferation of SEG-1 and BIC-1 esophageal cancer cells with half-maximal growth inhibitory concentration values of 36 and 48 muM, respectively. TA also increased Annexin V staining in both cell lines, indicative of proapoptotic activity. Treatment of SEG-1 and BIC-1 cells with TA for up to 72 h decreased expression of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 and this was accompanied by decreased expression of the well-characterized Sp-regulated genes cyclin D1, vascular endothelial growth factor and survivin. TA also decreased hepatocyte growth factor receptor, (c-Met), a receptor tyrosine kinase that is overexpressed in esophageal cancer cells and tumors and is an important drug target. Knockdown of Sp1, Sp3 and Sp4 by RNA interference in SEG-1 and BIC-1 cells also decreased c-Met expression, demonstrating that c-Met is an Sp-regulated gene in esophageal cancer cells. Sp1 was overexpressed in esophageal cancer cells and tumors and increased Sp1 staining was observed in esophageal tumors from patients. TA (20 mg/kg/day) also decreased tumor growth and weight in athymic nude mice bearing SEG-1 cells as xenografts and this was accompanied by increased apoptosis and decreased Sp1 and c-Met staining in tumors from treated mice. Thus, TA-dependent downregulation of Sp transcription factors and c-Met defines a novel chemotherapeutic approach for treatment of esophageal cancer.
Collapse
Affiliation(s)
- Sabitha Papineni
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
| | - Sudhakar Chintharlapalli
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, USA
| | - Maen Abdelrahim
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, FL 32806, USA
| | - Syng-ook Lee
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, USA
| | - Robert Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Ala Abudayyeh
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cheryl Baker
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, FL 32806, USA
| | - Luis Herrera
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, FL 32806, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, USA
| |
Collapse
|
34
|
How age affects the biology of breast cancer. Clin Oncol (R Coll Radiol) 2008; 21:81-5. [PMID: 19071000 DOI: 10.1016/j.clon.2008.11.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 11/17/2008] [Indexed: 01/21/2023]
Abstract
Breast cancer incidence increases with age, but there are important age-related differences with respect to the frequency of different tumour subtypes with respect to hormone receptor status and pathological grade. In general, younger patients show a higher frequency of oestrogen receptor-negative, higher-grade tumours, whereas in older patients there is a higher frequency of oestrogen receptor-positive, low-grade tumours. This accounts for the fact that, in general, elderly patients are thought to have a less aggressive form of the disease. However, this does not mean that all elderly patients with breast cancer necessarily have a good prognosis. An increased understanding of the mechanisms of tissue ageing and how these affect the molecular biological phenotype of breast cancers in cohorts of different ages will aid the oncologist's confidence in tailoring treatment more appropriately to the likely prognosis, and the development of novel, hopefully less toxic, treatments for specific subtypes of breast cancer in the elderly population.
Collapse
|
35
|
Thakur MK, Sharma PK. Binding of estrogen receptor alpha promoter to nuclear proteins of mouse cerebral cortex: effect of age, sex, and gonadal steroids. Biogerontology 2008; 9:467-78. [PMID: 18716892 DOI: 10.1007/s10522-008-9166-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 07/30/2008] [Indexed: 10/21/2022]
Abstract
Majority of estrogen actions in the brain are mediated by estrogen receptor (ER) alpha which in turn is regulated by several factors like circulating levels of gonadal steroid hormones 17beta-estradiol and testosterone, sex and age of the organism. The expression of ERalpha is regulated through interaction between cis-elements of its promoter and proteins present in the nuclei. Here, we have used electrophoretic mobility shift assay (EMSA) to analyze the effect of age, sex, 17beta-estradiol, and testosterone on the binding of ERalpha promoter (-91 to +46 bp) to nuclear proteins from the mouse cerebral cortex. EMSA revealed the formation of three specific complexes in all groups. However, the intensity of these complexes varied as a function of age, sex and treatment with 17beta-estradiol and testosterone. Nuclear proteins from the cerebral cortex of both sexes showed reduced binding with promoter fragment in old mice. Further, competition analysis indicated stronger binding in females than males of both ages. The extent of binding was reduced by 17beta-estradiol and testosterone treatment in both ages and sexes. Thus, these findings demonstrate differential binding of nuclear proteins to mouse ERalpha promoter which may account for different functions of estrogen in the brain.
Collapse
Affiliation(s)
- M K Thakur
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, India.
| | | |
Collapse
|
36
|
Thakur MK, Kumar RC. 17Beta-estradiol modulates age-dependent binding of 40 kDa nuclear protein to androgen receptor promoter in mouse cerebral cortex. Biogerontology 2007; 8:575-82. [PMID: 17619166 DOI: 10.1007/s10522-007-9102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 05/15/2007] [Indexed: 11/24/2022]
Abstract
Androgen influences the function of central and peripheral nervous system and plays a crucial role in maintaining reproductive behaviors and neuroendocrine regulation. Such action is mediated by interaction of androgen receptor (AR) promoter with nuclear proteins, which are involved in transcriptional regulation of androgen responsive genes. We have analyzed the binding of AR core promoter to nuclear proteins from the cerebral cortex of adult and old mice of both sexes by electrophoretic mobility shift assay (EMSA) and characterized the bound protein by Southwestern blotting. EMSA showed that the binding of nuclear proteins declined in the cerebral cortex of intact old mice as compared to adult. Following gonadectomy, the binding was reduced in old male and adult female but increased in old female. In contrast, estradiol supplementation increased the binding in old male and adult female but decreased in old female. Southwestern blotting analysis revealed that a 40 kDa nuclear protein bound to the promoter and the binding pattern was similar to that observed in EMSA. Further characterization of this protein may help to explore the intricate mechanism that underlies the transcriptional regulation of androgen responsive genes during aging.
Collapse
Affiliation(s)
- Mahendra K Thakur
- Biochemistry & Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| | | |
Collapse
|
37
|
Iannelli P, Zarrilli V, Varricchio E, Tramontano D, Mancini FP. The dietary antioxidant resveratrol affects redox changes of PPARalpha activity. Nutr Metab Cardiovasc Dis 2007; 17:247-256. [PMID: 17134953 DOI: 10.1016/j.numecd.2005.12.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 12/12/2005] [Accepted: 12/13/2005] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Gene-environment interaction is behind the pathogenesis of most widespread diseases, and nutrition is among the environmental factors with the highest impact on human health. The mechanisms involved in the interaction between nutritional factors and the genetic background of individuals are still unclear. The aim of this study was to investigate whether resveratrol (RES), an antioxidant polyphenol of red wine, can influence the activity of PPARalpha in the rat hepatoma cell line McArdle-RH7777. PPARalpha is a transcriptional factor that regulates gene expression when activated by endogenous or exogenous long-chain fatty acids. Its activation results in significant protection from cardiovascular diseases in humans. METHODS AND RESULTS By means of the electromobility shift assay (EMSA), we observed that PPARalpha is redox-sensitive as it displays reduced DNA-binding activity following in vivo treatment of the cells with 1mmol/L diethylmaleate (DEM), a glutathione-depleting agent. This finding could be relevant considering the important role of redox balance in pathological and physiological processes. We also observed a dual effect of 100mumol/L RES on PPARalpha activity: it was able to prevent, to a large extent, the DEM-induced reduction of DNA-binding activity at earlier time points, when the effect of DEM was stronger, but it depressed PPARalpha activity at later time points, when the effect of DEM was greatly reduced. CONCLUSION A nutritional substance, such as RES, is able to influence the activity of gene-regulating factors, but the net effect is difficult to predict when the compound involved has multiple biological properties. Caution is therefore warranted before drawing conclusions about the potential benefits of RES for human health.
Collapse
Affiliation(s)
- Paola Iannelli
- Department of Biological and Environmental Sciences, University of Sannio, via Port'Arsa 11, 82100 Benevento, Italy
| | | | | | | | | |
Collapse
|
38
|
Pathak RU, Kanungo MS. An age-specific 35-kDa phosphoprotein binds to a repressor element in the ovalbumin gene promoter in the avian species Japanese quail. DNA Cell Biol 2007; 26:44-54. [PMID: 17263596 DOI: 10.1089/dna.2006.0523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The ovalbumin (Ov) gene is expressed in the tubular gland cells of the avian oviduct in a specific manner under the control of developmental, tissue-specific, and hormonal cues. The expression is controlled by an array of positive and negative cis-acting elements present up to 1 kb upstream of its transcription start site. Our findings presented in this communication indicate that a well-characterized repressor element may be involved in active repression of the gene during aging. At least two proteins bind to the 25-bp sequence used in the present study encompassing the COUP adjacent repressor (CAR) element. The binding of one of the trans-acting factor that interacts with the repressor element increases during aging. This is accompanied by a decrease in transcription of the gene. The binding of the factor-to-repressor element decreases when expression of the Ov gene is induced by steroid administration. The factor has an approximate molecular weight of 35 kDa and is a phosphoprotein. It loses its ability to bind to DNA upon dephosphorylation. This makes it a potential target of various kinases/phosphatases that relay the various developmental, tissue-specific, and hormonal cues. The other trans-acting factor is a single-strand specific protein that interacts with the repressor element in an age-independent manner. These two proteins acting in conjunction may be involved in the repression of the Ov gene in old female birds where the lower circulating level of steroid hormones may be acting as an age-related cue.
Collapse
Affiliation(s)
- Rashmi U Pathak
- Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
39
|
Oh JE, Han JA, Hwang ES. Downregulation of transcription factor, Sp1, during cellular senescence. Biochem Biophys Res Commun 2006; 353:86-91. [PMID: 17161377 DOI: 10.1016/j.bbrc.2006.11.118] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 11/20/2006] [Indexed: 02/06/2023]
Abstract
We found that the protein level of Sp1 transcription factor decreases as normal human fibroblasts undergo replicative aging. Sp1 also undergoes a rapid decrease in the protein level and activity in MCF-7 cells that are induced to a state of cellular senescence. In the cells treated with other DNA damaging chemicals such as actinomycin D and H(2)O(2), the Sp1 level decreased progressively as well. Inhibition of ATM/ATR kinases prevented this downregulation, suggesting that DNA damage signaling is involved in the regulation of the Sp1. This decrease in Sp1 protein level is due to the accelerated proteasomal degradation since a proteasome inhibitor, ALLN, blocked this downregulation. Therefore, the global decrease in gene transcription frequently reported in aging cells and tissues could be attributed at least in part to the decrease in Sp1 level.
Collapse
Affiliation(s)
- Ji-Eun Oh
- Department of Life Science, University of Seoul, Dongdaemungu, Jeonnongdong 90, Seoul 130-743, Republic of Korea
| | | | | |
Collapse
|
40
|
Transcriptional and translational regulation of BACE1 expression--implications for Alzheimer's disease. Prog Neurobiol 2006; 79:95-111. [PMID: 16904810 DOI: 10.1016/j.pneurobio.2006.06.001] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 06/08/2006] [Accepted: 06/09/2006] [Indexed: 01/07/2023]
Abstract
The proteolytical processing of the amyloid precursor protein (APP) gives rise to beta-amyloid peptides, which accumulate in brains of Alzheimer's disease (AD) patients. Different soluble or insoluble higher molecular weight forms of beta-amyloid peptides have been postulated to trigger a complex pathological cascade that may cause synaptic dysfunction, inflammatory processes, neuronal loss, cognitive impairment, and finally the onset of the disease. The generation of beta-amyloid peptides requires the proteolytical cleavage of APP by an aspartyl protease named beta-site APP-cleaving enzyme 1 (BACE1). The expression and enzymatic activity of BACE1 are increased in brains of AD patients. Here we discuss the importance of a number of recently identified transcription factors as well as post-transcriptional modifications and activation of intracellular signaling molecules for the regulation of BACE1 expression in brain. Importantly, some of these factors are known to be involved in the inflammatory and chronic stress responses of the brain, which are compromised during aging. Moreover, recent evidence indicates that beneficial effects of non-steriodal anti-inflammatory drugs on the progression of AD are mediated--at least in part--by effects on the peroxisome proliferator-activated receptor-gamma response element present in the BACE1 promoter. The identification of the cell type-specific expression and activation of NF-kappaB, Sp1 and YY1 transcription factors may provide a basis to specifically interfere with BACE1 expression and, thereby, to lower the concentrations of beta-amyloid peptides, which may prevent neuronal cell loss and cognitive decline in AD patients.
Collapse
|
41
|
Musci G, Persichini T, Casadei M, Mazzone V, Venturini G, Polticelli F, Colasanti M. Nitrosative/oxidative modifications and ageing. Mech Ageing Dev 2006; 127:544-51. [PMID: 16530251 DOI: 10.1016/j.mad.2006.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2005] [Revised: 10/03/2005] [Accepted: 01/16/2006] [Indexed: 11/27/2022]
Abstract
We present here a brief description of the relationships among metals, nitric oxide metabolism, and ageing. In particular, we will discuss the interactions occurring between redox (copper, iron) and non-redox (zinc) metals and nitric oxide, the metal- and nitric oxide-catalyzed formation of thiol adducts (nitrosothiols, mixed disulphides) and the possible involvement of these species in the ageing process.
Collapse
Affiliation(s)
- Giovanni Musci
- Dipartimento di Scienze Microbiologiche, Genetiche e Molecolari, University of Messina, Italy
| | | | | | | | | | | | | |
Collapse
|
42
|
Jung KJ, Maruyama N, Ishigami A, Yu BP, Chung HY. The redox-sensitive DNA binding sites responsible for age-related downregulation of SMP30 by ERK pathway and reversal by calorie restriction. Antioxid Redox Signal 2006; 8:671-80. [PMID: 16677110 DOI: 10.1089/ars.2006.8.671] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
It was recently found that age-related changes in SMP30 expression can be modulated by antioxidative action. In the current study, the modulation of SMP30 gene expression was explored by (a) antioxidative calorie restriction (CR), (b) proinflammatory lipopolysaccharide (LPS), in aged rat, (c) oxidative stress promoter, tert-butylhydroperoxide (t-BHP)-injected mouse, and (d) t-BHP-treated Ac2F cells. Utilizing EMSA, particular attention was given to the binding activity of unidentified transcription factor in sites 3 and 5 that are located in -800 bp of the SMP30 promoter. Results showed that CR prevented the age-related decrease in SMP30 expression, and also showed that SMP30 gene expression and binding activities of sites 3 and 5 decreased with treatments of t-BHP or LPS. These findings were confirmed by the antioxidant NAC and ERK-specific inhibitor PD098059 that blunted decreased SMP30 gene expression and binding activity of sites 3 and 5 by t-BHP in Ac2F cell system. Our data strongly indicate that the SMP30 transcriptional process is redox-sensitive and its modulation occurs at DNA binding sites 3 and 5 in the promoter region. Perhaps a more significant finding of the present study is that the downregulation of SMP30 is likely involved in ERK signal pathway.
Collapse
Affiliation(s)
- Kyung Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | | | | | | | | |
Collapse
|
43
|
Ahn J, Ko M, Lee K, Oh J, Jeon SH, Seong RH. Expression of SRG3, a core component of mouse SWI/SNF chromatin-remodeling complex, is regulated by cooperative interactions between Sp1/Sp3 and Ets transcription factors. Biochem Biophys Res Commun 2005; 338:1435-46. [PMID: 16288722 DOI: 10.1016/j.bbrc.2005.10.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Accepted: 10/19/2005] [Indexed: 11/16/2022]
Abstract
SRG3, a mouse homolog of yeast SWI3 and human BAF155, is known to be a core component of SWI/SNF chromatin-remodeling complex. We have previously shown that SRG3 plays essential roles in early mouse embryogenesis, brain development, and T-cell development. SRG3 gene expression was differentially regulated depending on the developmental stages and exhibited tissue-specific pattern. In this study, we showed that the functional interactions between Sp and Ets family transcription factors are crucial for the SRG3 expression. Sp1 and Sp3 specifically bound to the two canonical Sp-binding sites (GC boxes) at -152 and -114, and a non-canonical Sp-binding site (CCTCCT motif) at -108 in the SRG3 promoter. Using Drosophila SL2 cells, we found that various Sp or Ets family members activate the SRG3 promoter through these Sp- or Ets-binding sites, respectively, in a dose-dependent manner. Intriguingly, different combinatorial expression of Ets and Sp factors in SL2 cells resulted in either strong synergistic activation or repression of the SRG3 promoter activity. Moreover, the Sp-mediated activation of SRG3 promoter required the intact Ets-binding element. Taken together, these results suggest that diverse interactions between Sp1/Sp3 and Ets factors are crucial for the SRG3 gene expression.
Collapse
Affiliation(s)
- Jeongeun Ahn
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Research Center for Functional Cellomics, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | |
Collapse
|
44
|
Mocchegiani E, Bertoni-Freddari C, Marcellini F, Malavolta M. Brain, aging and neurodegeneration: role of zinc ion availability. Prog Neurobiol 2005; 75:367-90. [PMID: 15927345 DOI: 10.1016/j.pneurobio.2005.04.005] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2004] [Revised: 04/20/2005] [Accepted: 04/29/2005] [Indexed: 10/25/2022]
Abstract
Actual fields of research in neurobiology are not only aimed at understanding the different aspects of brain aging but also at developing strategies useful to preserve brain compensatory capacity and to prevent the onset of neurodegenerative diseases. Consistent with this trend much attention has been addressed to zinc metabolism. In fact, zinc acts as a neuromodulator at excitatory synapses and has a considerable role in the stress response and in the functionality of zinc-dependent enzymes contributing to maintaining brain compensatory capacity. In particular, the mechanisms that modulate the free zinc pool are pivotal for safeguarding brain health and performance. Alterations in zinc homeostasis have been reported in Parkinson's and Alzheimer's disease as well as in transient forebrain ischemia, seizures and traumatic brain injury, but little is known regarding aged brain. There is much evidence that that age-related changes, frequently associated to a decline in brain functions and impaired cognitive performances, could be related to dysfunctions affecting the intracellular zinc ion availability. A general agreement emerges from studies of humans' and rodents' old brains about an increased expression of metallothionein (MT) isoforms I and II, but dyshomogenous results are reported for MT-III, and it is still uncertain whether these proteins maintain in aging the protective role, as it occurs in adult/young age. At the same time, there is considerable evidence that amyloid-beta deposition in Alzheimer's disease is induced by zinc, but the pathological significance and the causes of this phenomenon are still an open question. The scientific debate on the role of zinc and of some zinc-binding proteins in aging and neurodegenerative disorders, as well as on the beneficial effect of zinc supplementation in aged brain and neurodegeneration, is extensively discussed in this review.
Collapse
Affiliation(s)
- Eugenio Mocchegiani
- Immunology Ctr. Section Nutrition, Immunity and Aging, Res. Department INRCA, Ancona 60100, Via Birarelli 8, 60121, Italy.
| | | | | | | |
Collapse
|
45
|
Tsuda M, Watanabe T, Seki T, Kimura T, Sawa H, Minami A, Akagi T, Isobe KI, Nagashima K, Tanaka S. Induction of p21WAF1/CIP1 by human synovial sarcoma-associated chimeric oncoprotein SYT-SSX1. Oncogene 2005; 24:7984-90. [PMID: 16103879 DOI: 10.1038/sj.onc.1208942] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Oncogenic protein provokes cell cycle arrest termed premature senescence. In this process Ras has been known to induce cyclin-dependent kinase inhibitor (CKI) p16(INK4A) in primary fibroblasts. Here, we present a novel finding that human chimeric oncoprotein SYT-SSX1 induces CKI p21(WAF1/CIP1) (p21) for suppression of cell growth. In human synovial sarcoma cell lines, the expression levels of p21 were high and the transcriptional activity of the p21 gene promoter was significantly elevated. The transient expression of SYT-SSX1-induced activation of the p21 gene promoter in human diploid fibroblasts. The N-terminus deletion form of SYT-SSX1, which failed to bind to hBRM one of the chromatin remodeling factors, preserved the p21 induction ability. This effect of SYT-SSX1 was similar in extent in both wild-type and p53-deficient HCT116 cell lines. Furthermore, the introduction of mutation in Sp1/Sp3 binding sites of the p21 gene promoter abolished the SYT-SSX1-induced transcriptional activity of its promoter. In SW13 cells, the stable expression of SYT-SSX1 suppressed cell growth in culture. These results suggest that SYT-SSX1 is able to induce p21 in a manner independent on hBRM and p53 but dependent on Sp1/Sp3.
Collapse
Affiliation(s)
- Masumi Tsuda
- Laboratory of Molecular and Cellular Pathology, Hokkaido University Graduate School of Medicine, N15, W7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Jiang Y, Wang L, Gong W, Wei D, Le X, Yao J, Ajani J, Abbruzzese JL, Huang S, Xie K. A high expression level of insulin-like growth factor I receptor is associated with increased expression of transcription factor Sp1 and regional lymph node metastasis of human gastric cancer. Clin Exp Metastasis 2005; 21:755-64. [PMID: 16035620 DOI: 10.1007/s10585-005-1198-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor I receptor (IGF-IR) is critical to cell survival and growth and altered IGF-IR expression is found in many human cancers. However, its expression and potential role in gastric cancer development and progression has not been explored. The IGF-IR expression level was determined via immunohistochemistry in primary tumor and lymph node metastasis of 86 cases of resected gastric cancer. Relationships of IGF-IR expression with transcription factor Spl expression and clinicopathological features were analyzed. The impact of altered Sp1 expression on IGF-IR expression and gastric cancer biology was further determined using small inhibitory RNA for Sp1 mRNA. We found that IGF-IR was overexpressed in 62% of the tumor samples when compared with adjacent tumor-free gastric mucosa. Patients with lymph node metastases had strong expression of IGF-IR in both primary and metastatic tumor cells. IGF-IR overexpression in the primary tumor correlated with increased lymph node metastasis. Furthermore, the level of IGF-IR expression directly correlated with that of Spl, an important transcription factor for IGF-IR regulation. Knocking-down of Spl expression by small inhibitory RNA led to decreased IGF-IR expression and attenuated growth and metastasis of gastric cancer cells. Therefore, dysregulated expression of IGF-IR and/or Sp1 may contribute to the growth and metastasis of gastric cancer and potentially can be a target of therapeutic intervention.
Collapse
Affiliation(s)
- Yixing Jiang
- Department of Gastrointestinal Medical Oncology, The University of Texas, MD, Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Zinc (Zn) deficiency, a frequent condition in human populations, induces oxidative stress and subsequently activates/inhibits oxidant-sensitive transcription factors that can affect cell function, proliferation and survival leading to disease. Zn deficiency-triggered oxidative stress could affect cell signaling, including: (1) transcription factors containing Zn finger motifs, and (2) other oxidant-sensitive transcription factors (NF-kappaB and AP-1). The Zn finger motif in the Zn finger transcription factors is mainly a DNA binding domain. Cysteine residues coordinate the Zn ion folding structural domains that participate in intermolecular interactions. Oxidative stress can impair the DNA-binding activity of Zn finger transcription factor, by oxidizing the cysteine residues and therefore altering the secondary structure of the protein. AP-1 is generally activated in Zn deficiency that can occur secondary to an increase in cellular H(2)O(2), followed by activation of MAPKs p38 and JNK. The role of AP-1 in Zn deficiency-associated pathology remains to be established. The cytosolic steps of the NF-kappaB cascade are activated by oxidants in Zn deficiency. However, an impaired nuclear transport of the active transcription factor leads to a low expression of NF-kappaB-dependent genes that could be involved in multiple aspects of Zn deficiency associated pathology. In summary, Zn deficiency induces oxidative stress that can both, lead to tissue oxidative damage and/or to the modulation of select signaling cascades. Their role in the pathology of Zn deficiency remains to be defined.
Collapse
Affiliation(s)
- Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
48
|
Zhang C, Wang J, Cheng J, Liu G, Chen Q. Seeking for senile-related gene expression in cerebral tissue of senescence-accelerated mouse. Cell Mol Neurobiol 2004; 24:741-7. [PMID: 15672676 PMCID: PMC11529920 DOI: 10.1007/s10571-004-6915-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. A better understanding of the molecular effect on aging in the brain may help reveal important aspects of organism aging, as well as the processes that lead to aging-related brain dysfunction. In this study, the aging-specific expression genes of the murine cerebrum were investigated by using the technique of DDRT-PCR in two senescence-accelerated mouse strains, SAMP10/Ta and SAMR1TA. 2. Through comparing gene expression profile among the age, 2, 4, 12, and 18 month of the SAMP10/Ta strain, four differential fragments have been found, and comparing gene expression profile between the two mouse strains, 24 fragments have been detected, 7 and 17 of them belong to SAMP10/Ta and SAMR1TA, respectively. 3. Sequencing analysis indicated that most of those fragments are homologous with some of certain gene cDNA that are related with senile. The data obtained from this study suggest that many genes are involved in the senile process and accelerate senescence phenotypic pathologies in SAMP10/Ta.
Collapse
Affiliation(s)
- Chong Zhang
- The Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- The Graduate School of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jingang Wang
- Lingtai Bicheng Medical Technology Co. Ltd., Beijing, People’s Republic of China
| | - Jinyan Cheng
- The Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Guisheng Liu
- The Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Qingxuan Chen
- The Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
49
|
Abstract
Cognitive aspects of aging represent a grave challenge for our societal circumstances as members of the baby-boom generation spiral toward a collective 'senior moment'. In addition, age-related changes in the CNS can contribute to motor deficits and other somatic aberrations. Inflammation and its regulation by cytokines have been connected to many aspects of aging, and mechanisms addressed here provide a rationale for this. Nevertheless, a role for cytokines in normal aging of the human brain has not been confirmed, and it seems to be possible to ameliorate both cognitive decline and cytokine elevation via lifestyle choices. So ignorance of the brain should not prohibit development of successful strategies for delaying or avoiding neurological deficits.
Collapse
Affiliation(s)
- Angela M Bodles
- Donald W. Reynolds Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | |
Collapse
|
50
|
Quong J, Eppenberger-Castori S, Moore D, Scott GK, Birrer MJ, Kueng W, Eppenberger U, Benz CC. Age-dependent changes in breast cancer hormone receptors and oxidant stress markers. Breast Cancer Res Treat 2002; 76:221-36. [PMID: 12462383 DOI: 10.1023/a:1020886801674] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer incidence increases with age but this relationship has not been fully explored with regard to expression of estrogen receptor (ER) and ER-inducible genes (PR, pS2, Bcl2, cathepsin D), or the age-dependence of oxidant stress markers that also affect ER-inducible gene expression. In this three-part study, we first correlated age at diagnosis with expression of breast cancer markers ER, PR, pS2, Bcl2, and cathepsin D, quantitated by enzyme immunoassays from a European collective of approximately 3000 cryobanked primary breast cancers and approximately 300 adjacent non-malignant breast tissues. Results were then compared with ER and PR data reported to the SEER registry for 83,541 US cancers diagnosed during 1992-1997. Lastly, a homogeneous subset of 70 ER-positive tumors preselected from the European collective was blindly analyzed for age-specific changes in the DNA-binding content of redox-sensitive transcriprtion factors, AP1 and Sp1, and the oxidant stress-activated protein kinase, phosphorylated(P)-Erk5. Increases in breast tumor ER from patients aged <30 to >80 years mirrored 10-fold lower increases in non-malignant breast tissue ER content up to age 60, rising faster thereafter and reaching a near 25-fold differential between malignant and non-malignant breast tissue by age 80. ER-inducible markers PR, pS2, Bcl2, and cathepsin D were overexpressed in tumors relative to non-malignant breast tissue but, unlike ER, did not increase with patient age. While SEER data demonstrated that the increase in US breast cancer incidence rates after age 50 is confined to ER-positive tumors in patients of all ethnic subsets, these patients also showed a striking increase in the proportion of higher-risk ER-positive/PR-negative breast cancers arising after age 50. Mechanistically essential for ER-inducible PR expression, Sp1 DNA-binding function (but not Sp1 content) was lost with age in ER-positive tumors; and this functional defect correlated with increased tumor content of the oxidant stress marker, P-Erk5. Altogether these findings support two hypotheses: (i) dysregulated ER expression underlies the age-specific increase in breast cancer incidence after age 50; and (ii) oxidative stress and loss of Sp1 DNA-binding may contribute to an increasing incidence in higher-risk ER-positive/PR-negative breast cancers with aging.
Collapse
Affiliation(s)
- Judy Quong
- Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|