20 results on '"Shao, Z."'
Search Results
2. MiRNA-621 sensitizes breast cancer to chemotherapy by suppressing FBXO11 and enhancing p53 activity
- Author
-
Xue, J, Chi, Y, Chen, Y, Huang, S, Ye, X, Niu, J, Wang, W, Pfeffer, L M, Shao, Z-m, Wu, Z-H, and Wu, J
- Published
- 2016
- Full Text
- View/download PDF
3. Inhibition of EGFR pathway signaling and the metastatic potential of breast cancer cells by PA-MSHA mediated by type 1 fimbriae via a mannose-dependent manner
- Author
-
Liu, Z-B, Hou, Y-F, Zhu, J, Hu, D-L, Jin, W, Ou, Z-L, Di, G-H, Wu, J, Shen, Z-Z, and Shao, Z-M
- Published
- 2010
- Full Text
- View/download PDF
4. Downregulation of miR-193b contributes to enhance urokinase-type plasminogen activator (uPA) expression and tumor progression and invasion in human breast cancer
- Author
-
Li, X-F, Yan, P-J, and Shao, Z-M
- Published
- 2009
- Full Text
- View/download PDF
5. Enhanced expression of Duffy antigen receptor for chemokines by breast cancer cells attenuates growth and metastasis potential
- Author
-
Wang, J, Ou, Z-L, Hou, Y-F, Luo, J-M, Shen, Z-Z, Ding, J, and Shao, Z-M
- Published
- 2006
- Full Text
- View/download PDF
6. Induction of miRNA-181a by genotoxic treatments promotes chemotherapeutic resistance and metastasis in breast cancer
- Author
-
Niu, J, primary, Xue, A, additional, Chi, Y, additional, Xue, J, additional, Wang, W, additional, Zhao, Z, additional, Fan, M, additional, Yang, C H, additional, Shao, Z-M, additional, Pfeffer, L M, additional, Wu, J, additional, and Wu, Z-H, additional
- Published
- 2015
- Full Text
- View/download PDF
7. MiRNA-621 sensitizes breast cancer to chemotherapy by suppressing FBXO11 and enhancing p53 activity
- Author
-
Xue, J, primary, Chi, Y, additional, Chen, Y, additional, Huang, S, additional, Ye, X, additional, Niu, J, additional, Wang, W, additional, Pfeffer, L M, additional, Shao, Z-m, additional, Wu, Z-H, additional, and Wu, J, additional
- Published
- 2015
- Full Text
- View/download PDF
8. The Ku70-SIX1-GPT2 axis regulates alpha-ketoglutarate metabolism to drive progression of prostate cancer.
- Author
-
Huang H, Zhuang X, Yin S, Sun W, Cheng J, Peng EY, Xiang Y, He X, Tang M, Li Y, Yao Y, Deng Y, Liu Q, Shao Z, Xia X, Cai G, and Liao Y
- Abstract
Sine oculis homeobox homolog 1 (SIX1) is a new identified cancer driver in the development of prostate cancer (PC). However, the upstream regulatory mechanisms for SIX1 reactivation in cancer remains elusive. Here, we found that Ku70 robustly interacts with SIX1 in the nucleus of PC cells. The HD domain of SIX1 and the DBD domain of Ku70 are required for formation of Ku70-SIX1 complex. 20 groups of hydrogen bonds were identified in this complex by molecular dynamics simulation. Depletion of Ku70/SIX1 notably abrogates the proliferation and migration of PC. Further studies revealed that SIX1 is recruited to the promoter region on glutamate-pyruvate transaminase 2 (GPT2). Ku70 enhances the SIX1-mediated transcriptional activation on GPT2, thereby facilitating the generation of alpha-ketoglutarate (α-KG). In addition, formation of the Ku70-SIX1 complex promotes GPT2-dependent cell proliferation and migration in PC. Moreover, the expression of GPT2 is upregulated and strongly correlated with the expression of Ku70/SIX1 in PC tissues. In summary, our findings not only provide insight into the mechanistic interactions between Ku70 and SIX1, but also highlight the significance of the Ku70-SIX1-GPT2 axis for α-KG metabolism and PC carcinogenesis., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
9. NNMT switches the proangiogenic phenotype of cancer-associated fibroblasts via epigenetically regulating ETS2/VEGFA axis.
- Author
-
Wang X, Zhao H, Luo X, Chen Y, Shi C, Wang Y, Bai J, Shao Z, and Shang Z
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Phenotype, Signal Transduction genetics, Tumor Microenvironment genetics, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Neovascularization, Pathologic genetics, Neovascularization, Pathologic pathology, Neovascularization, Pathologic metabolism, Nicotinamide N-Methyltransferase genetics, Nicotinamide N-Methyltransferase metabolism, Proto-Oncogene Protein c-ets-2 genetics, Proto-Oncogene Protein c-ets-2 metabolism, Squamous Cell Carcinoma of Head and Neck genetics, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A genetics, Mouth Neoplasms genetics, Mouth Neoplasms metabolism, Mouth Neoplasms pathology
- Abstract
Cancer-associated fibroblasts (CAFs) are known to promote angiogenesis in oral squamous cell carcinoma (OSCC). However, the epigenetic mechanisms through which CAFs facilitate angiogenesis within the tumor microenvironment are still poorly characterized. Nicotinamide N'-methyltransferase (NNMT), a member of the N-methyltransferase family, was found to be a key molecule in the activation of CAFs. This study shows that NNMT in fibroblasts contributes to angiogenesis and tumor growth through an epigenetic reprogramming-ETS2-VEGFA signaling axis in OSCC. Single-cell RNA Sequencing (scRNA-seq) analysis suggests that NNMT is mainly highly expressed in fibroblasts of head and neck squamous cell carcinoma (HNSCC). Moreover, analysis of the TCGA database and multiple immunohistochemical staining of clinical samples also identified a positive correlation between NNMT and tumor angiogenesis. This research further employed an assembled organoid model and a fibroblast-endothelial cell co-culture model to authenticate the proangiogenic ability of NNMT. At the molecular level, high expression of NNMT in CAFs was found to promote ETS2 expression by regulating H3K27 methylation level through mediating methylation deposition. Furthermore, ETS2 was verified to be an activating transcription factor of VEGFA in this study. Collectively, our findings delineate an epigenetic molecular regulatory network of angiogenesis and provide a theoretical basis for exploring new targets and clinical strategy in OSCC., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
10. KLHL29-mediated DDX3X degradation promotes chemosensitivity by abrogating cell cycle checkpoint in triple-negative breast cancer.
- Author
-
Yao L, Hao Q, Wang M, Chen Y, Cao H, Zhang Q, Yu K, Jiang Y, Shao Z, Zhou X, and Xu Y
- Subjects
- Humans, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cell Proliferation genetics, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases metabolism, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology
- Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous breast cancer subtype and accounts for approximately 15-20% of breast cancer cases. In this study, we identified KLHL29, which is an understudied member of the Kelch-like gene family, as a crucial tumor suppressor that regulates chemosensitivity in TNBC. KLHL29 expression was significantly downregulated in breast cancer tissues compared with adjacent normal tissues, and low levels of KLHL29 were associated with unfavorable prognoses. Ectopic KLHL29 suppressed, while depleting KLHL29 promoted, the growth, proliferation, migration, and invasion of TNBC. Mechanistically, KLHL29 recruited the CUL3 E3-ligase to the RNA-binding protein DDX3X, leading to the proteasomal degradation of the latter. This downregulation of DDX3X resulted in the destabilization of CCND1 mRNA and the consequent cell cycle arrest at G0/G1 phase. Remarkably, the DDX3X inhibitor RK33 combined with platinum-based chemotherapy can synergistically suppress TNBC that usually expresses low levels of KLHL29 and high levels of DDX3X using cancer cell-derived xenograft and patient-derived organoids models. Altogether, we uncovered the potential role for the KLHL29-DDX3X signaling cascade in the regulation of TNBC progression, thus providing a promising combination strategy for overcoming TNBC chemoresistance., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. Autophagy regulates the cancer stem cell phenotype of head and neck squamous cell carcinoma through the noncanonical FOXO3/SOX2 axis.
- Author
-
Chen Y, Zhao H, Liang W, Jiang E, Zhou X, Shao Z, Liu K, and Shang Z
- Subjects
- Humans, Animals, Cell Line, Tumor, Mice, Phenotype, Gene Expression Regulation, Neoplastic, Carcinoma, Squamous Cell pathology, Carcinoma, Squamous Cell metabolism, Carcinoma, Squamous Cell genetics, Signal Transduction, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, SOXB1 Transcription Factors metabolism, SOXB1 Transcription Factors genetics, Autophagy genetics, Forkhead Box Protein O3 metabolism, Forkhead Box Protein O3 genetics, Squamous Cell Carcinoma of Head and Neck pathology, Squamous Cell Carcinoma of Head and Neck metabolism, Squamous Cell Carcinoma of Head and Neck genetics, Head and Neck Neoplasms pathology, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms genetics
- Abstract
Autophagy is an essential catabolic process that orchestrates cellular homeostasis and plays dual roles in tumor promotion and suppression. However, the mechanism by which autophagy affects the self-renewal of cancer stem cells (CSCs) remains unclear. In this study, we investigated whether autophagy activation contributes to CSC properties of head and neck squamous cell carcinoma (HNSCC). The results showed that the autophagy level and CSC properties of HNSCC cells were elevated in response to several adverse conditions, including treatment with cisplatin, starvation, and hypoxia. Pretreatment with autophagy inhibitors, such as 3-MA and chloroquine, diminished the CSC properties acquired under adverse conditions. In addition, the isolated CSCs were endowed with stronger autophagic activity than non-CSCs, and the CSC properties were dampened when autophagy was inhibited either by 3-MA, chloroquine, or Beclin1 knockdown. Notably, the tumor-initiating activity of CSCs was decreased upon knocking down Beclin1. Further study revealed that FOXO3, a substrate for autophagy, was enriched in the nucleus of cells with lower autophagy levels. Nuclear FOXO3 directly bound to the promoter region of SOX2 and negatively regulated its transcriptional activity. Overexpression of FOXO3 decreased the expression of SOX2 and thereby impaired the CSC phenotype both in vitro and in vivo. Taken together, our findings suggest that the activation of autophagy is essential for the acquisition of CSC properties in adverse conditions and the self-renewal of CSCs. We clarify the role of autophagy in regulating the CSC phenotype and demonstrate that the noncanonical FOXO3/SOX2 axis is the intrinsic regulatory mechanism., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. A new role of GRP75-USP1-SIX1 protein complex in driving prostate cancer progression and castration resistance.
- Author
-
Liao Y, Liu Y, Shao Z, Xia X, Deng Y, Cai J, Yao L, He J, Yu C, Hu T, Sun W, Liu F, Tang D, Liu J, and Huang H
- Subjects
- Animals, Castration, Cell Line, Tumor, Cell Proliferation genetics, Disease Progression, Gene Expression Regulation, Neoplastic genetics, HEK293 Cells, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Nude, PC-3 Cells, Prostate pathology, Prostatic Neoplasms, Castration-Resistant pathology, Proteolysis, Receptors, Androgen genetics, Signal Transduction genetics, Transcription Factors genetics, Ubiquitination genetics, HSP70 Heat-Shock Proteins genetics, Homeodomain Proteins genetics, Mitochondrial Proteins genetics, Prostatic Neoplasms, Castration-Resistant genetics, Ubiquitin-Specific Proteases genetics
- Abstract
Prostate cancer (PC) is the second most common cancer with limited treatment option in males. Although the reactivation of embryonic signals in adult cells is one of the characteristics of cancer, the underlying protein degradation mechanism remains elusive. Here, we show that the molecular chaperone GRP75 is a key player in PC cells by maintaining the protein stability of SIX1, a transcription factor for embryonic development. Mechanistically, GRP75 provides a platform to recruit the deubiquitinating enzyme USP1 to inhibit K48-linked polyubiquitination of SIX1. Structurally, the C-terminus of GRP75 (433-679 aa) contains a peptide binding domain, which is required for the formation of GRP75-USP1-SIX1 protein complex. Functionally, pharmacological or genetic inhibition of the GRP75-USP1-SIX1 protein complex suppresses tumor growth and overcomes the castration resistance of PC cells in vitro and in xenograft mouse models. Clinically, the protein expression of SIX1 in PC tumor tissues is positively correlated with the expression of GRP75 and USP1. These new findings not only enhance our understanding of the protein degradation mechanism, but also may provide a potential way to enhance the anti-cancer activity of androgen suppression therapy.
- Published
- 2021
- Full Text
- View/download PDF
13. BCL9/BCL9L promotes tumorigenicity through immune-dependent and independent mechanisms in triple negative breast cancer.
- Author
-
Wang X, Feng M, Xiao T, Guo B, Liu D, Liu C, Pei J, Liu Q, Xiao Y, Rosin-Arbesfeld R, Shi Y, Zhou Y, Yang M, Feng YX, Jiang Y, Shao Z, Yu K, and Zhu D
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Cell Proliferation physiology, Humans, MCF-7 Cells, Mice, Mice, Nude, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Tumor Microenvironment, DNA-Binding Proteins immunology, Transcription Factors immunology, Triple Negative Breast Neoplasms immunology
- Abstract
Treatment of patients with triple-negative breast cancer (TNBC) has been challenging due to a lack of well-defined molecular targets. The Wnt/β-catenin pathway is known to be activated in many TNBC patients and BCL9 and BCL9L are important transcriptional co-activators of β-catenin, but whether inhibition of BCL9/BCL9L can suppress TNBC growth and the underlying mechanism are not fully understood. Here we demonstrate that the expression of BCL9 and BCL9L is directly correlated with malignancy in TNBC patient tumors and that BCL9 and BCL9L promote tumor cell growth, cell migration and metastasis in TNBC models. Mechanistically, we found that BCL9/BCL9L promotes tumorigenicity through both the Wnt and TGF-β pathways. Besides, BCL9/BCL9L expression inversely correlates with CD8
+ T cell infiltration in TNBC and BCL9/BCL9L inhibits the infiltration of CD8+ T cells in the tumor microenvironment. hsBCL9CT -24, an inhibitor of BCL9/β-catenin peptides, promotes intratumoral infiltration of cytotoxic T cells, reducing regulatory T cells (Treg) and increasing dendritic cells (DCs). Inhibition of BCL9/BCL9L and TGF-β suppresses activity of Treg. TGF-β signaling increases tumor infiltration of cytotoxic CD8+ T cells. In accordance, genetic or pharmacological inhibition of BCL9/BCL9L synergizes with PD-1/L1 antibodies to inhibit tumor growth. In summary, these results suggest that targeting BCL9/BCL9L has a direct anti-tumor effect and also unleashes an anti-cancer immune response through inhibition of both Wnt and TGF-β signaling, suggesting a viable therapeutic approach for TNBC treatment.- Published
- 2021
- Full Text
- View/download PDF
14. Human breast carcinoma desmoplasia is PDGF initiated.
- Author
-
Shao ZM, Nguyen M, and Barsky SH
- Subjects
- Animals, Breast pathology, Breast Neoplasms metabolism, Carcinogenicity Tests, Carcinoma metabolism, Collagen metabolism, Estradiol pharmacology, Fibroblasts pathology, Gene Expression Regulation, Neoplastic drug effects, Genes, ras, Humans, Insulin-Like Growth Factor II drug effects, Insulin-Like Growth Factor II genetics, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Matrix Metalloproteinase 11, Metalloendopeptidases drug effects, Metalloendopeptidases genetics, Mice, Mice, Nude, Mutation, Platelet-Derived Growth Factor genetics, Tissue Inhibitor of Metalloproteinase-1 drug effects, Tissue Inhibitor of Metalloproteinase-1 genetics, Transplantation, Heterologous, Tumor Cells, Cultured, Breast Neoplasms pathology, Carcinoma pathology, Platelet-Derived Growth Factor metabolism
- Abstract
The desmoplastic response to human breast carcinoma is a host myofibroblast-mediated collagenous response exhibiting synergistic effects on tumor progression. Although many paracrine interactions between breast carcinoma cells and myofibroblasts have been characterized, the event(s) which initiate desmoplasia have remained undefined. Our studies utilized c-rasH transfected MCF-7 cells which overexpress ras p2l and which are weakly tumorigenic in ovariectomized nude mice. The xenografts are desmoplastic and comprised of 30% myofibroblasts and 60 mg/g of interstitial collagen. In situ hybridization studies of these xenografts reveal a stromal gene expression pattern (stromelysin-3, IGF-II and TIMP-1) identical to that observed in human tumor desmoplasia. 17-beta estradiol increases c-rasH MCF-7 growth but abolishes desmoplasia. c-rasH MCF-7 in vitro constitutively produce myofibroblast mitogenic activity which competes with PDGF in a receptor binding assay. This myofibroblast mitogenic activity is unaltered by 17-beta estradiol/tamoxifen pretreatment in vitro. Transfection of c-rasH MCF-7 with a PDGF-A dominant negative mutant, 1308, produced by site-directed mutagenesis (serine-->cysteine129) reduces both homo- and heterodimer secretion of PDGF by as much as 90% but does not interfere with the secretion of other growth factors. Clones with low PDGF, though tumorigenic, are non-desmoplastic. Our results suggest that breast carcinoma-secreted PDGF is the major initiator of tumor desmoplasia.
- Published
- 2000
- Full Text
- View/download PDF
15. The human myoepithelial cell displays a multifaceted anti-angiogenic phenotype.
- Author
-
Nguyen M, Lee MC, Wang JL, Tomlinson JS, Shao ZM, Alpaugh ML, and Barsky SH
- Subjects
- Animals, Cell Hypoxia, Cell Line, Culture Media, Conditioned pharmacology, Culture Media, Serum-Free pharmacology, Endothelial Growth Factors biosynthesis, Endothelial Growth Factors genetics, Endothelium, Vascular cytology, Epithelial Cells pathology, Extracellular Matrix metabolism, Female, Gene Expression Regulation, Neoplastic, Growth Substances biosynthesis, Growth Substances genetics, Humans, Lymphokines biosynthesis, Lymphokines genetics, Mice, Mice, Nude, Mice, SCID, Necrosis, Neoplasm Metastasis, Neoplasm Proteins genetics, Nitric Oxide Synthase biosynthesis, Nitric Oxide Synthase genetics, Nitric Oxide Synthase Type II, Phenotype, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Neoplasm biosynthesis, RNA, Neoplasm genetics, Receptors, Fibroblast Growth Factor genetics, Thrombospondin 1 genetics, Tissue Inhibitor of Metalloproteinase-1 genetics, Transplantation, Heterologous, Tumor Cells, Cultured metabolism, Tumor Cells, Cultured pathology, Umbilical Veins cytology, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Epithelial Cells metabolism, Neoplasm Proteins biosynthesis, Neovascularization, Pathologic, Receptors, Fibroblast Growth Factor biosynthesis, Thrombospondin 1 biosynthesis, Tissue Inhibitor of Metalloproteinase-1 biosynthesis
- Abstract
Human myoepithelial cells which surround ducts and acini of certain organs such as the breast form a natural border separating epithelial cells from stromal angiogenesis. Myoepithelial cell lines (HMS-1-6), derived from diverse benign myoepithelial tumors, all constitutively express high levels of active angiogenic inhibitors which include TIMP-1, thrombospondin-1 and soluble bFGF receptors but very low levels of angiogenic factors. These myoepithelial cell lines inhibit endothelial cell chemotaxis and proliferation. These myoepithelial cell lines sense hypoxia, respond to low O2 tension by increased HIF-1 alpha but with only a minimal increase in VEGF and iNOS steady state mRNA levels. Their corresponding xenografts (HMS-X-6X) grow very slowly compared to their non-myoepithelial carcinomatous counterparts and accumulate an abundant extracellular matrix devoid of angiogenesis but containing bound angiogenic inhibitors. These myoepithelial xenografts exhibit only minimal hypoxia but extensive necrosis in comparison to their non-myoepithelial xenograft counterparts. These former xenografts inhibit local and systemic tumor-induced angiogenesis and metastasis presumably from their matrix-bound and released circulating angiogenic inhibitors. These observations collectively support the hypothesis that the human myoepithelial cell (even when transformed) is a natural suppressor of angiogenesis. Oncogene (2000) 19, 3449 - 3459
- Published
- 2000
- Full Text
- View/download PDF
16. Cyclin D1 associates with the TBP-associated factor TAF(II)250 to regulate Sp1-mediated transcription.
- Author
-
Adnane J, Shao Z, and Robbins PD
- Subjects
- 3T3 Cells, Animals, Cell Line, Cyclin D1 genetics, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Histone Acetyltransferases, Humans, Mice, Mutagenesis, Nuclear Proteins genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Retinoblastoma Protein genetics, Retinoblastoma Protein metabolism, Sp1 Transcription Factor genetics, Spodoptera, TATA-Box Binding Protein, Transcription, Genetic, Cyclin D1 metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Gene Expression Regulation, Nuclear Proteins metabolism, Proto-Oncogene Proteins, Sp1 Transcription Factor metabolism, TATA Box, TATA-Binding Protein Associated Factors, Transcription Factor TFIID, Transcription Factors
- Abstract
We have previously shown that Sp1-mediated transcription is stimulated by Rb and repressed by cyclin D1. The stimulation of Sp1 transcriptional activity by Rb is conferred, in part, through a direct interaction with the TBP-associated factor TAF(II)250. Here we investigated the mechanism(s) through which cyclin D1 represses Sp1. We examined the ability of cyclin D1 to regulate transcription mediated by Gal4-Sp1 fusion proteins, which contain the Gal4 DNA-binding domain and Sp1 trans-activation domain(s). The domain of Sp1 sufficient to confer repression by cyclin D1 was mapped to a region important for interaction with TAF(II)110. We further demonstrate that TAF(II)250-cyclin D1 complexes can be immunoprecipitated from mammalian and baculovirus-infected insect cells and that recombinant GST-TAF(II)250 (amino acids 1-434) associates with cyclin D1 in vitro. Moreover, the overexpression of Rb or CDK4 reduced the level of TAF(II)250-cyclin D1 complex. The amino terminus of cyclin D1 (amino acids 1-100) was sufficient for association with TAF(II)250 and for repressing Sp1-mediated transcription. Taken together, the results suggest that cyclin D1 may regulate transcription by interacting directly or indirectly with TAF(II)250.
- Published
- 1999
- Full Text
- View/download PDF
17. Rb interacts with TAF(II)250/TFIID through multiple domains.
- Author
-
Shao Z, Siegert JL, Ruppert S, and Robbins PD
- Subjects
- Binding Sites, HeLa Cells, Humans, Transcription Factor TFIID, Retinoblastoma Protein metabolism, Transcription Factors, TFII metabolism
- Abstract
The retinoblastoma tumor suppressor gene product (Rb) binds directly to the largest TFIID subunit, TATA-binding protein associated factor TAF(II)250, first identified as the cell cycle regulatory protein CCG1. Here we map the domains in Rb and TAF(II)250 important for their interaction in vitro and in vivo. Both the amino terminus and the large pocket of Rb are able to associate independently with TAF(II)250. The binding domain(s) within the large pocket are distinct from the viral oncoprotein and E2F binding region since certain pocket mutations, which abolish E1A binding, do not abolish TAF(II)250 binding. Consistent with the large pocket of Rb binding to TAF(II)250, the large pocket domains of both p107 and p130 are able to bind to TAF(II)250 in vivo. We also demonstrate that at least two regions of TAF(II)250 are able to bind to the large pocket of Rb independently whereas the amino terminus of Rb binds to a distinct domain in TAF(II)250. We further demonstrate that Rb can bind to TFIID in vitro, presumably in part through an interaction with TAF(II)250. Our results suggest a complex interaction between Rb and TAF(II)250 and imply that TAF(II)250, TFIID, and potentially other basal transcription factors are targets for regulation by Rb and Rb-related proteins.
- Published
- 1997
- Full Text
- View/download PDF
18. p53 independent G0/G1 arrest and apoptosis induced by a novel retinoid in human breast cancer cells.
- Author
-
Shao ZM, Dawson MI, Li XS, Rishi AK, Sheikh MS, Han QX, Ordonez JV, Shroot B, and Fontana JA
- Subjects
- Cyclin-Dependent Kinase Inhibitor p21, Cyclins genetics, Cyclins metabolism, DNA Damage, Gene Expression Regulation, Neoplastic drug effects, Humans, In Vitro Techniques, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2, RNA, Messenger genetics, Transcription Factor AP-1 metabolism, Transcriptional Activation, Tumor Cells, Cultured, Tumor Suppressor Protein p53 physiology, bcl-2-Associated X Protein, Apoptosis drug effects, Breast Neoplasms pathology, Cell Cycle drug effects, Growth Inhibitors, Naphthalenes pharmacology, Retinoids pharmacology
- Abstract
The biological activity of a novel synthetic retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (AHPN) was investigated in human breast carcinoma (HBC) cells. Although capable of selective binding to the RAR gamma nuclear receptor, AHPN inhibited the growth of a number of HBC cell lines via RAR- or RXR-independent pathways. AHPN also inhibited the growth of the human leukemia cell line HL-60R which does not possess functional RARs. RA significantly inhibited AP-1 mediated gene activation in MCF-7 cells while AHPN displayed no such anti-AP-1 activity. Retinoids normally are cytostatic in their inhibition of breast carcinoma growth and permit cell proliferation upon their removal, wher as AHPN induced G0/G1 arrest within 6h followed by apoptosis. In MCF-7 cells that harbor wild type p53, AHPN-induced G0/G1 arrest and apoptosis was accompanied by p53-independent regulation of WAF1/CIP1 as well as bax mRNA levels while bcl-2 mRNA levels were decreased. In MDA-MB-231 cells which possess a mutant p53, AHPN-mediated G0/G1 arrest and apoptosis was also associated with a concomitant up regulation of WAF1/CIP1 mRNA while these cells did not express bax or bcl-2 messages. Thus AHPN represents a novel retinoid that induces G0/G1 arrest and apoptosis via a unique pathway which appears to involve activation of known downstream effectors of p53 in a p53-independent manner.
- Published
- 1995
19. Differential regulation of E2F and Sp1-mediated transcription by G1 cyclins.
- Author
-
Shao Z and Robbins PD
- Subjects
- 3T3 Cells, Animals, Cyclins genetics, Mice, Promoter Regions, Genetic, Transcription, Genetic drug effects, Transfection, Adenovirus E2 Proteins pharmacology, Cyclins pharmacology, Gene Expression Regulation, Neoplastic, Retinoblastoma Protein metabolism, Sp1 Transcription Factor pharmacology
- Abstract
Cyclins have been demonstrated to mediate phosphorylation of the retinoblastoma tumor suppressor gene product (Rb) and/or to bind directly to Rb. Since Rb is a regulator of E2F and Sp1-mediated transcription, we have examined the effect of overexpression of cyclins on transcription mediated by E2F-dependent adenovirus E2 promoter and by Sp1 in a cotransfection assay in 3T3 cells. All the G1 cyclins tested, C, D1, D2, D3 and E, as well as cyclin A were able to stimulate E2 promoter activity to various levels with D3 showing the strongest stimulation. For stimulation of the E2 promoter by cyclins A, E and D-type cyclins was dependent upon the presence of functional E2F and ATF binding sites. Cyclin C, however, was able to stimulate both E2F and ATF-dependent transcription to the same level as the wild type E2 promoter. In addition, cyclin C was able to stimulate transcription mediated by Sp1, GAL4-Sp1 and GAL4-VP16, suggesting that cyclin C affects a general pathway of transcriptional activation. In contrast, cyclin D1 was able to repress specifically Sp1-mediated transcription through an Rb-independent pathway. These results suggest that cyclins can regulate transcription mediated by specific transcription factors in both positive and negative manners. Furthermore, the results demonstrate clear functional differences between the G1 cyclins, in particular, functional differences between the related D-type cyclins.
- Published
- 1995
20. Mechanisms of regulation of WAF1/Cip1 gene expression in human breast carcinoma: role of p53-dependent and independent signal transduction pathways.
- Author
-
Sheikh MS, Li XS, Chen JC, Shao ZM, Ordonez JV, and Fontana JA
- Subjects
- Cell Cycle genetics, Cyclin-Dependent Kinase Inhibitor p21, Down-Regulation, Humans, Promoter Regions, Genetic, Protein Kinase Inhibitors, RNA, Messenger genetics, RNA, Messenger metabolism, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Breast Neoplasms genetics, Cyclins genetics, Gene Expression Regulation, Neoplastic genetics, Signal Transduction, Tumor Suppressor Protein p53 physiology
- Abstract
WAF1/Cip1 was recently identified as the wild-type p53 target that appears to mediate the tumor suppressing effects of p53. We investigated the mechanisms of regulation of WAF1/Cip1 gene expression in human breast carcinoma (HBC) cells. Our results demonstrate that the HBC cells harboring wild-type p53 express 26-33-fold higher WAF1/Cip1 mRNA levels than the cells harboring mutant p53. The DNA damaging agent etoposide induced p53 accumulation only in cells harboring wild-type p53 yet it induced WAF1/Cip1 gene expression in cells carrying wild-type or mutant p53, suggesting the involvement of p53-dependent and independent signaling pathways in the regulation of WAF1/Cip1 gene expression. Serum starvation-induced growth arrest although not altering the endogenous p53 levels or its ability to transactivate the reporter gene, induced WAF1/Cip1 gene expression in cells carrying wild-type as well as mutant p53. These results further implicated the involvement of p53-independent signal transduction pathways in WAF1/Cip1 gene regulation. Our data also suggest that WAF1/Cip1 gene expression is tightly associated with cell cycle progression in cells containing either wild-type or mutant p53. WAF1/Cip1 expression was transiently induced in response to serum treatment and declined as the cells passed through the S-phase of the cell cycle. We thus provide evidence that the mechanisms of WAF1/Cip1 gene regulation involve p53-dependent and independent signaling pathways in HBC.
- Published
- 1994
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.