1
|
Liu T, Du J, Cheng X, Wei J. Integrative Analysis of the Role of TP53 in Human Pan-Cancer. Curr Issues Mol Biol 2023; 45:9606-9633. [PMID: 38132447 PMCID: PMC10742156 DOI: 10.3390/cimb45120601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
Tumor protein P53 (TP53) is an important tumor suppressor gene in humans. Under normal circumstances, TP53 can help repair mutated genes, or promote the death of cells with severe gene mutations (specifically, TP53 prevents cells from arrest in the G1/S phase when deoxyribonucleic acid (DNA) is damaged and promotes apoptosis if not repaired), and prevents normal cells from becoming malignant cells. TP53 mutations affect its tumor suppressor function, leading to the development of malignant tumors. In this study, using a public database, we explored the pan-cancer expression of TP53, its impact on patient survival and prognosis, the types of gene mutations, its correlation with immunity, and its regulation of other transcription factors and micro RNA (miRNA). The docking sites of therapeutic drugs and key amino acid sites of action provide a basis for future targeted therapies. TP53 has important biological functions in the human body. This study provides a theoretical basis for clinical TP53 gene therapy.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; (T.L.); (J.D.)
| | - Jin Du
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; (T.L.); (J.D.)
| | - Xiangshu Cheng
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; (T.L.); (J.D.)
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; (T.L.); (J.D.)
| |
Collapse
|
2
|
Babu D, Chintal R, Panigrahi M, Phanithi PB. Distinct expression and function of breast cancer metastasis suppressor 1 in mutant P53 glioblastoma. Cell Oncol (Dordr) 2022; 45:1451-1465. [PMID: 36284039 DOI: 10.1007/s13402-022-00729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2022] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Glioblastoma (GBM) is the most malignant subtype of astrocytic tumors with the worst prognosis in all its progressive forms. Breast cancer metastasis suppressor 1 (BRMS1) is a metastasis suppressor gene that controls malignancy in multiple tumors. As yet, however, its clinical and functional significance in mutant P53 GBM remains inconclusive. Here, we attempted to study the importance of BRMS1 in mutant P53 GBM. METHODS BRMS1 expression was evaluated in 74 human astrocytoma tissues by qRT-PCR, Western blotting and immunohistochemistry. BRMS1 expression in the astrocytoma tissues was correlated with clinicopathological parameters, the P53 mutation status and BRMS1 downstream targets, and compared with TCGA and NCI-60 datasets. siRNA-mediated knockdown of BRMS1 was performed in selected GBM cell lines to evaluate the functional role of BRMS1. RESULTS Our study revealed an enhanced expression of BRMS1 in GBM which was associated with a poor patient survival, and this observation was corroborated by the TCGA dataset. We also found a positive correlation between BRMS1 expression and a mutant P53 status in GBM which was associated with a poor prognosis. In vitro BRMS1 silencing reduced the growth of mutant P53 GBM cells and repressed their colonization and migration/invasion by modulating EGFR-AKT/NF-κB signaling. Transcriptional profiling revealed a positive and negative correlation of uPA and ING4 expression with BRMS1 expression, respectively. CONCLUSION Our data indicate upregulation of BRMS1 in high grade astrocytomas which correlates positively with mutant P53 and a poor patient survival. Silencing of BRMS1 in mutant P53 GBM cell lines resulted in a reduced cellular growth and migration/invasion by suppressing the EGFR-AKT/NF-kB signaling pathway. BRMS1 may serve as a predictive biomarker and therapeutic target in mutant P53 GBM.
Collapse
Affiliation(s)
- Deepak Babu
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India
| | - Ramulu Chintal
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India
| | - Manas Panigrahi
- Department of Neurosurgery, Krishna Institute of Medical Sciences, 500 003, Secunderabad, Telangana State, India
| | - Prakash Babu Phanithi
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India.
| |
Collapse
|
3
|
Shah S, Pendleton E, Couture O, Broachwalla M, Kusper T, Alt LAC, Fay MJ, Chandar N. P53 regulation of osteoblast differentiation is mediated through specific microRNAs. Biochem Biophys Rep 2021; 25:100920. [PMID: 33553686 PMCID: PMC7859171 DOI: 10.1016/j.bbrep.2021.100920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/17/2022] Open
Abstract
In order to understand the role of the p53 tumor suppressor gene in microRNA expression during osteoblast differentiation, we used a screen to identify microRNAs that were altered in a p53-dependent manner. MicroRNAs from MC3T3-E1 preosteoblasts were isolated from day 0 (undifferentiated) and day 4 (differentiating) and compared to a p53 deficient MC3T3-E1 line treated similarly. Overall, one fourth of all the microRNAs tested showed a reduction of 0.6 fold, and a similar number of them were increased 1.7 fold with differentiation. P53 deficiency caused 40% reduction in expression of microRNAs in differentiating cells, while a small percent (0.03%) showed an increase. Changes in microRNAs were validated using real-time PCR and two microRNAs were selected for further analysis (miR-34b and miR-140). These two microRNAs were increased significantly during differentiation but showed a dramatic reduction in expression in a p53 deficient state. Stable expression of miR-34b and miR-140 in MC3T3-E1 cells resulted in decreases in cell proliferation rates when compared to control cells. There was a 4-fold increase in p53 levels with miR-34b expression and a less dramatic increase with miR-140. Putative target binding sites for bone specific transcription factors, Runx2 and Osterix, were found for miR-34b, while Runx2, beta catenin and type 1 collagen were found to be miR-140 targets. Western blot analyses and functional assays for the transcription factors Runx2, Osterix and Beta-catenin confirmed microRNA specific interactions. These studies provide evidence that p53 mediated regulation of osteoblast differentiation can also occur through specific microRNAs such as miR-34b and miR-140 that also directly target important bone specific genes. The p53 tumor suppressor gene regulates microRNA expression during in vitro osteoblast differentiation. miR34b and miR140 targets include several bone specific markers such as runx2, beta catenin, type 1 collagen and osterix. miR34b and miR140 overexpression inhibits osteoblast cell proliferation.
Collapse
Affiliation(s)
- Shivang Shah
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Elisha Pendleton
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Oliver Couture
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Mustafa Broachwalla
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Teresa Kusper
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Lauren A C Alt
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Michael J Fay
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA.,Department of Pharmacology, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| | - Nalini Chandar
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 555, 31st, Street, Downers Grove, IL60515, USA
| |
Collapse
|
4
|
IER2-induced senescence drives melanoma invasion through osteopontin. Oncogene 2021; 40:6494-6512. [PMID: 34611309 PMCID: PMC8616759 DOI: 10.1038/s41388-021-02027-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 09/01/2021] [Accepted: 09/17/2021] [Indexed: 01/07/2023]
Abstract
Expression of the immediate-early response gene IER2 has been associated with the progression of several types of cancer, but its functional role is poorly understood. We found that increased IER2 expression in human melanoma is associated with shorter overall survival, and subsequently investigated the mechanisms through which IER2 exerts this effect. In experimental melanoma models, sustained expression of IER2 induced senescence in a subset of melanoma cells in a p53/MAPK/AKT-dependent manner. The senescent cells produced a characteristic secretome that included high levels of the extracellular phosphoglycoprotein osteopontin. Nuclear localization of the IER2 protein was critical for both the induction of senescence and osteopontin secretion. Osteopontin secreted by IER2-expressing senescent cells strongly stimulated the migration and invasion of non-senescent melanoma cells. Consistently, we observed coordinate expression of IER2, p53/p21, and osteopontin in primary human melanomas and metastases, highlighting the pathophysiological relevance of IER2-mediated senescence in melanoma progression. Together, our study reveals that sustained IER2 expression drives melanoma invasion and progression through stimulating osteopontin secretion via the stochastic induction of senescence.
Collapse
|
5
|
Gómez‐Santos B, Saenz de Urturi D, Nuñez‐García M, Gonzalez‐Romero F, Buque X, Aurrekoetxea I, Gutiérrez de Juan V, Gonzalez‐Rellan MJ, García‐Monzón C, González‐Rodríguez Á, Mosteiro L, Errazti G, Mifsut P, Gaztambide S, Castaño L, Martin C, Nogueiras R, Martinez‐Chantar ML, Syn W, Aspichueta P. Liver osteopontin is required to prevent the progression of age-related nonalcoholic fatty liver disease. Aging Cell 2020; 19:e13183. [PMID: 32638492 PMCID: PMC7431823 DOI: 10.1111/acel.13183] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/14/2020] [Accepted: 06/06/2020] [Indexed: 12/17/2022] Open
Abstract
Osteopontin (OPN), a senescence-associated secretory phenotype factor, is increased in patients with nonalcoholic fatty liver disease (NAFLD). Cellular senescence has been associated with age-dependent hepatosteatosis. Thus, we investigated the role of OPN in the age-related hepatosteatosis. For this, human serum samples, animal models of aging, and cell lines in which senescence was induced were used. Metabolic fluxes, lipid, and protein concentration were determined. Among individuals with a normal liver, we observed a positive correlation between serum OPN levels and increasing age. This correlation with age, however, was absent in patients with NAFLD. In wild-type (WT) mice, serum and liver OPN were increased at 10 months old (m) along with liver p53 levels and remained elevated at 20m. Markers of liver senescence increased in association with synthesis and concentration of triglycerides (TG) in 10m OPN-deficient (KO) hepatocytes when compared to WT hepatocytes. These changes in senescence and lipid metabolism in 10m OPN-KO mice liver were associated with the decrease of 78 kDa glucose-regulated protein (GRP78), induction of ER stress, and the increase in fatty acid synthase and CD36 levels. OPN deficiency in senescent cells also diminished GRP78, the accumulation of intracellular TG, and the increase in CD36 levels. In 20m mice, OPN loss led to increased liver fibrosis. Finally, we showed that OPN expression in vitro and in vivo was regulated by p53. In conclusion, OPN deficiency leads to earlier cellular senescence, ER stress, and TG accumulation during aging. The p53-OPN axis is required to inhibit the onset of age-related hepatosteatosis.
Collapse
Affiliation(s)
- Beatriz Gómez‐Santos
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
| | - Diego Saenz de Urturi
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
| | - Maitane Nuñez‐García
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
| | - Francisco Gonzalez‐Romero
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
| | - Xabier Buque
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Igor Aurrekoetxea
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Virginia Gutiérrez de Juan
- Liver Disease Lab, Center for Cooperative Research in Bioscience (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) e Derio Bizkaia Spain
| | - Maria J. Gonzalez‐Rellan
- Department of Physiology CIMUS University of Santiago de Compostela‐Instituto de Investigación Sanitaria Santiago de Compostela Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
| | - Carmelo García‐Monzón
- Liver Research Unit Santa Cristina University Hospital Instituto de Investigación Sanitaria Princesa Madrid Spain
- Centro de investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute Madrid Spain
| | - Águeda González‐Rodríguez
- Liver Research Unit Santa Cristina University Hospital Instituto de Investigación Sanitaria Princesa Madrid Spain
- Centro de investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute Madrid Spain
| | - Lorena Mosteiro
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Gaizka Errazti
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Patricia Mifsut
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Sonia Gaztambide
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Luis Castaño
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| | - Cesar Martin
- Department of Biochemistry and Molecular Biology Biofisika Institute (UPV/EHU, CSIC) UPV/EHU Spain
| | - Rubén Nogueiras
- Department of Physiology CIMUS University of Santiago de Compostela‐Instituto de Investigación Sanitaria Santiago de Compostela Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
| | - María L. Martinez‐Chantar
- Liver Disease Lab, Center for Cooperative Research in Bioscience (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) e Derio Bizkaia Spain
- Centro de investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute Madrid Spain
| | - Wing‐Kin Syn
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
- Section of Gastroenterology Ralph H Johnson VAMC Charleston SC USA
- Division of Gastroenterology and Hepatology Medical University of South Carolina Charleston SC USA
| | - Patricia Aspichueta
- Department of Physiology Faculty of Medicine and Nursing University of Basque Country UPV/EHU Leioa Spain
- Biocruces Bizkaia Health Research Institute Cruces University Hospital Barakaldo Spain
| |
Collapse
|
6
|
Koyama R, Tamura M, Nakagaki T, Ohashi T, Idogawa M, Suzuki H, Tokino T, Sasaki Y. Identification and characterization of a metastatic suppressor BRMS1L as a target gene of p53. Cancer Sci 2017; 108:2413-2421. [PMID: 29030916 PMCID: PMC5715288 DOI: 10.1111/cas.13420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor p53 and its family members, p63 and p73, play a pivotal role in the cell fate determination in response to diverse upstream signals. As transcription factors, p53 family proteins regulate a number of genes that are involved in cell cycle arrest, apoptosis, senescence, and maintenance of genomic stability. Recent studies revealed that p53 family proteins are important for the regulation of cell invasion and migration. Microarray analysis showed that breast cancer metastasis suppressor 1‐like (BRMS1L) is upregulated by p53 family proteins, specifically p53, TAp63γ, and TAp73β. We identified two responsive elements of p53 family proteins in the first intron and upstream of BRMS1L. These response elements are well conserved among mammals. Functional analysis showed that ectopic expression of BRMS1L inhibited cancer cell invasion and migration; knockdown of BRMS1L by siRNA induced the opposite effect. Importantly, clinical databases revealed that reduced BRMS1L expression correlated with poor prognosis in patients with breast and brain cancer. Together, these results strongly indicate that BRMS1L is one of the mediators downstream of the p53 pathway, and that it inhibits cancer cell invasion and migration, which are essential steps in cancer metastasis. Collectively, our results indicate that BRMS1L is involved in cancer cell invasion and migration, and could be a therapeutic target for cancer.
Collapse
Affiliation(s)
- Ryota Koyama
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Miyuki Tamura
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Takafumi Nakagaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Tomoko Ohashi
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Masashi Idogawa
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University, Sapporo, Japan
| | - Takashi Tokino
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Yasushi Sasaki
- Department of Medical Genome Sciences, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
7
|
Seidel SD, Stott WT, Kan HL, Sparrow BR, Gollapudi BB. Gene Expression Dose-Response of Liver with a Genotoxic and Nongenotoxic Carcinogen. Int J Toxicol 2016; 25:57-64. [PMID: 16510358 DOI: 10.1080/10915810500488429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tumorigenic mechanisms due to chemical exposure are broadly classified as either genotoxic or nongenotoxic. Genotoxic mechanisms are generally well defined; however nongenotoxic modes of tumorgenesis are less straightforward. This study was undertaken to help elucidate dose-response changes in gene expression (transcriptome) in the liver of rats in response to administration of known genotoxic or nongenotoxic liver carcinogens. Male Big Blue Fischer 344 rats were treated for 28-days with 0, 0.1, 0.3, 1.0, or 3.0 mg/kg/day of the genotoxin 2-acetylaminofluorene (AAF) or 0, 10, 30, 60, or 100 mg/kg/day of the nongenotoxin phenobarbital (PB). Transcriptome analysis was performed using the relatively focused Clontech Rat Toxicology II microarray (465 genes) and hybridized with 32P-labeled cDNA target. The analysis indicated that after 28 days of treatment, AAF altered the expression of 14 genes (9 up-and 5 down-regulated) and PB altered the expression of 18 genes (10 up- and 8 down-regulated). Of the limited genes whose expression was altered by AAF and PB, four were altered in common, two up-regulated, and two down-regulated. Several of the genes that show modulation of transcriptional activity following AAF and PB treatment display an atypical dose-response relationship such that the expression at the higher doses tended to be similar to that of control. This high-dose effect could potentially be caused by adaptation, toxicity, or tissue remodeling. These results suggest that the transcriptional response of the cells to higher doses of a toxic agent is likely to be different from that of a low-dose exposure.
Collapse
Affiliation(s)
- Shawn D Seidel
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan 48674, USA.
| | | | | | | | | |
Collapse
|
8
|
Naciff JM, Daston GP. Toxicogenomic Approach to Endocrine Disrupters: Identification of a Transcript Profile Characteristic of Chemicals with Estrogenic Activity. Toxicol Pathol 2016; 32 Suppl 2:59-70. [PMID: 15503665 DOI: 10.1080/01926230490463812] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Public concerns have been raised in recent years over the possible adverse effects that may result from exposure to chemicals in the environment that have the potential to interfere with the normal function of the endocrine system in wildlife and humans (“endocrine disrupters”). Regulations have been established that require the testing of pesticides used in food crops and drinking water contaminants, for estrogenicity and other hormonal activities. In the United States, the U.S. EPA proposed the Endocrine Disrupter Screening Program, which consists of a Tier 1 screening battery of tests that is designed to identify chemicals capable of interacting with various hormonal systems, and different Tier 2 testing assays that are designed to verify and broaden the Tier 1 results. We identify 2 main problems with this approach: (1) the fact that the developmental stages that are the most susceptible to endocrine disruption are not represented in the screening tier, mainly because developmental effects tend to be latent, and there is no way to economically screen in developing models; and (2) the expense to screen each chemical to be included in this program. Thus, the need arises for an accurate, rapid, and cost effective method for assessing the potential endocrine activity of multiple chemicals during development. We hypothesize that the largely latent developmental effects of some endocrine disruptors are preceded by immediate changes in gene expression in the embryo and fetus. Therefore, an approach to assess the potential estrogenic (and other steroid hormonal) activity of different compounds is to identify those patterns of gene expression elicited in a tissue/organ exposed to these particular classes of chemicals. In this paper, the potential utility of such an approach for screening and better understanding of mechanism of action for specific chemicals with endocrine disrupter activities is presented, using as an example chemicals with estrogenic activity.
Collapse
Affiliation(s)
- Jorge M Naciff
- Miami Valley Laboratories, The Procter and Gamble Company, Cincinnati, Ohio 45253, USA.
| | | |
Collapse
|
9
|
Peng G, Dan W, Jun W, Junjun Y, Tong R, Baoli Z, Yang X. Transcriptome profiling of the cancer and adjacent nontumor tissues from cervical squamous cell carcinoma patients by RNA sequencing. Tumour Biol 2015; 36:3309-17. [PMID: 25586346 DOI: 10.1007/s13277-014-2963-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/08/2014] [Indexed: 01/28/2023] Open
Abstract
Cervical cancer is the third most common cancer and the fourth leading cause of cancer deaths among women in the world. The discovery of vital diagnostic and therapeutic markers against cervical squamous cell carcinoma (CSCC) would broaden our understanding on the molecular basis of CSCC. In this study, we thoroughly analyzed the transcriptome of CSCC and matched adjacent nontumor (ATN) tissue. RNA sequencing was performed to screen the differentially expressed genes (DEGs) of three pairs of CSCC and ATN tissues. Functional enrichment analysis was used to uncover the biological functions of DEGs. Protein interaction network was carried out to reveal interaction of DEGs. Quantitative real-time PCR was conducted to validate the expression of DEGs. Immunohistochemistry was used to detect the relationship between clinicopathological parameters of CSCC and DEGs. There were a total of 347 significantly common DEGs in the three paired examples, including 104 consistent upregulated and 148 consistent downregulated DEGs. The 347 DEGs were categorized into 73 functional categories by Gene Ontology (GO) analysis. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis suggested six significantly signal pathways. The protein interaction network uncovered three important DEGs, including retinol dehydrogenase 12 (RDH12), ubiquitin D (UBD), and serum amyloid A1 (SAA1). We found that RDH12 expression was decreased in 74.5 % of CSCC tissues. RDH12 expression was negatively associated with tumor size and depth of cervical invasion. The UBD was overexpressed in 61.7 % of CSCC tissues and was positively related with tumor size and lymphatic metastasis. The SAA1 protein was overexpressed in 57.4 % of CSCC tissues and was positively related with clinicopathological parameters of tumor size, lymphatic metastasis, and depth of cervical invasion. The RDH12, UBD, and SAA1 genes might participate in the progression of CSCC.
Collapse
Affiliation(s)
- Guo Peng
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, No. 1 Shuai Fu Yuan, Wang Fu Jing Street, Beijing, 100730, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
10
|
Couture O, Lombardi E, Davis K, Hays E, Chandar N. Gene expression profiles resulting from stable loss of p53 mirrors its role in tissue differentiation. PLoS One 2013; 8:e82494. [PMID: 24312426 PMCID: PMC3842970 DOI: 10.1371/journal.pone.0082494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/25/2013] [Indexed: 01/24/2023] Open
Abstract
The tumor suppressor gene p53 is involved in a variety of cellular activities such as cellular stress responses, cell cycle regulation and differentiation. In our previous studies we have shown p53’s transcription activating role to be important in osteoblast differentiation. There is still a debate in the literature as to whether p53 inhibits or promotes differentiation. We have found p53 heterozygous mice to show a p53 dependency on some bone marker gene expression that is absent in knockout mice. Mice heterozygous for p53 also show a higher incidence of osteosarcomas than p53 knockout mice. This suggests that p53 is able to modify the environment within osteoblasts. In this study we compare changes in gene expression resulting after either a transient or stable reduction in p53. Accordingly we reduced p53 levels transiently and stably in C2C12 cells, which are capable of both myoblast and osteoblast differentiation, and compared the changes in gene expression of candidate genes regulated by the p53 pathway. Using a PCR array to assay for p53 target genes, we have found different expression profiles when comparing stable versus transient knockdown of p53. As expected, several genes with profound changes after transient p53 loss were related to apoptosis and cell cycle regulation. In contrast, stable p53 loss produced a greater change in MyoD and other transcription factors with tissue specific roles, suggesting that long term loss of p53 affects tissue homeostasis to a greater degree than changes resulting from acute loss of p53. These differences in gene expression were validated by measuring promoter activity of different pathway specific genes involved in differentiation. These studies suggest that an important role for p53 is context dependent, with a stable reduction in p53 expression affecting normal tissue physiology more than acute loss of p53.
Collapse
Affiliation(s)
- Oliver Couture
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States of America
| | - Eric Lombardi
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States of America
| | - Kendra Davis
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States of America
| | - Emily Hays
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States of America
| | - Nalini Chandar
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Illinois, United States of America
- * E-mail:
| |
Collapse
|
11
|
Pazolli E, Alspach E, Milczarek A, Prior J, Piwnica-Worms D, Stewart SA. Chromatin remodeling underlies the senescence-associated secretory phenotype of tumor stromal fibroblasts that supports cancer progression. Cancer Res 2012; 72:2251-61. [PMID: 22422937 DOI: 10.1158/0008-5472.can-11-3386] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Age is a major risk factor for the development of cancer. Senescent fibroblasts, which accumulate with age, secrete protumorigenic factors collectively referred to as the senescence-associated secretory phenotype (SASP). Here, we examined the molecular mechanisms that control SASP activation, focusing on the known SASP factor osteopontin (OPN). We found that expression of the canonical SASP members interleukin (IL)-6 and IL-8, but not OPN, were dependent upon a persistent DNA damage response (DDR) as evidenced by ATM and NF-κB activation. Treatment with several histone deacetylase (HDAC) inhibitors robustly activated SASP in the absence of DNA breaks, suggesting that DDR-dependent SASP activation occurs in response to chromatin remodeling rather than physical breaks in DNA. In the setting of HDAC inhibition, IL-6 and IL-8 expression remained dependent upon ATM and NF-κB, while OPN expression remained independent of these factors. Further analysis revealed that HDAC1 inhibition was sufficient to induce OPN expression, which is interesting given that loss of HDAC1 expression correlates with increased OPN expression within the stromal compartment of invasive breast cancers. Importantly, fibroblasts treated with HDAC inhibitors promoted tumor growth in vivo. Our findings therefore indicate that HDAC modulation plays an important role in stromal cell activation, with important implications for the use of HDAC inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Ermira Pazolli
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
12
|
Chen H, Kolman K, Lanciloti N, Nerney M, Hays E, Robson C, Chandar N. p53 and MDM2 are involved in the regulation of osteocalcin gene expression. Exp Cell Res 2012; 318:867-76. [PMID: 22405968 DOI: 10.1016/j.yexcr.2012.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 11/19/2022]
Abstract
Osteocalcin (OC) is a major noncollagenous bone matrix protein and an osteoblast marker whose expression is limited to mature osteoblasts during the late differentiation stage. In previous studies we have shown osteosarcomas to lose p53 function with a corresponding loss of osteocalcin gene expression. Introduction of wild type p53 resulted in re expression of the osteocalcin gene. Using gel shift and chromatin immunoprecipitation assays, we have identified a putative p53 binding site within the rat OC promoter region and observed an increase in OC promoter activity when p53 accumulates using a CAT assay. The p53 inducible gene Mdm2 is a well-known downstream regulator of p53 levels. Our results showed a synergistic increase in the OC promoter activity when both p53 and MDM2 were transiently overexpressed. We further demonstrate that p53 is not degraded during overexpression of MDM2 protein. Increased OC expression was observed with concomitantly increased p53, VDR, and MDM2 levels in ROS17/2.8 cells during treatment with differentiation promoting (DP) media, but was significantly decreased when co-treated with DP media and the small molecule inhibitor of MDM2-p53 interaction, Nutlin-3. We have also observed a dramatic increase of the OC promoter activity in the presence of p53 and Mdm2 with inclusion of Cbfa-1 and p300 factors. Our results suggest that under some physiological conditions the oncoprotein MDM2 may cooperate with p53 to regulate the osteocalcin gene during osteoblastic differentiation.
Collapse
Affiliation(s)
- Hankui Chen
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Li J, Yang GZ, Zhu ZM, Zhou ZY, Li L. Osteopontin is overexpressed in colorectal carcinoma and is correlated with P53 by immunohistochemistry. Exp Ther Med 2012; 3:621-624. [PMID: 22969940 DOI: 10.3892/etm.2012.465] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/17/2012] [Indexed: 12/27/2022] Open
Abstract
Osteopontin (OPN), a secreted phosphorylated glycoprotein, has been found to be involved in carcinogenesis, progression and metastasis of several types of cancers. The aim of the present study was to investigate the immunohistochemical expression of OPN in colorectal carcinoma (CRC) and its relationship with clinicopathological parameters and P53. Expression of OPN, Ki-67 and TP53 was detected in 77 cases of CRC by immunohistochemistry and the correlation of the expression of OPN with clinicopathological features, Ki-67 and P53 staining was investigated. Thirty-eight cases (49.4%) of CRC demonstrated OPN overexpression. Overexpression of OPN was associated with lymph node metastasis (P=0.025) and Dukes' stages (P=0.031), but not with gender, histological differentiation, depth of tumor invasion, TNM stages or Ki-67 index. The correlation between expression of OPN and TP53 was statistically significant (P=0.030). In conclusion, OPN is overexpressed in CRC, and plays a role in tumor progression and metastasis, which is possibly regulated by P53.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, the General Hospital of Beijing Military Command, Beijing 100700
| | | | | | | | | |
Collapse
|
14
|
Chen H, Hays E, Liboon J, Neely C, Kolman K, Chandar N. Osteocalcin gene expression is regulated by wild-type p53. Calcif Tissue Int 2011; 89:411-8. [PMID: 21964930 DOI: 10.1007/s00223-011-9533-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/03/2011] [Indexed: 01/14/2023]
Abstract
The tumor-suppressor p53 is a transcription factor that regulates a number of genes in the process of cell-cycle inhibition, apoptosis, and DNA damage. Recent studies have revealed a crucial role for p53 in bone remodeling. In our previous studies we have shown that p53 is an important regulator of osteoblast differentiation. In this study we investigated the role of p53 in the regulation of human osteocalcin gene expression. We observed that osteocalcin promoter activity could be upregulated by both exogenous and endogenous p53 and downregulated by p53-specific small interfering RNA. DNA affinity immunoblotting assay showed that p53 can bind to the human osteocalcin promoter in vitro. We further identified a p53 response element within the osteocalcin promoter region using a chromatin immunoprecipitation assay. Furthermore, we observed an additive effect of p53 and VDR on the regulation of osteocalcin promoter activity. Our findings suggest that p53 may directly target the human osteocalcin gene and positively affect osteocalcin gene expression.
Collapse
Affiliation(s)
- Hankui Chen
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | | | | | | | | | | |
Collapse
|
15
|
Chen C, Fu X, Zhang D, Li Y, Xie Y, Li Y, Huang Y. Varied pathways of stage IA lung adenocarcinomas discovered by integrated gene expression analysis. Int J Biol Sci 2011; 7:551-66. [PMID: 21552421 PMCID: PMC3088877 DOI: 10.7150/ijbs.7.551] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 03/31/2011] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Discovery of the progression-associated genes and pathways in lung adenocarcinoma (LAD) has important implications in understanding the molecular mechanism of tumor development. However, few studies had been performed to focus on the changes of pathways in lung adenocarcinoma development using microarray expression profile. RESULT We performed a meta-analysis of 4 LAD microarray datasets encompassing 353 patients to reveal differentially expressed genes (DEGs) between normal lung tissues and LAD of different stages. Overall, 1 838 genes were found to be dys-regulated, and the adipogenesis, circadian rhythm, and Id pathways were significantly changed. Interestingly, most of the genes from the same gene family (such as Interleukin receptor, Matrix metallopeptidase, Histone cluster and Minichromosome maintenance complex component families) were found to be up-regulated (or down-regulated). Real-time PCR (qRT-PCR) was applied to validate the expression of randomly selected 18 DEGs in LAD cell lines. In the pathway analysis among stages, Oxidative stress, Glycolysis/Gluconeogenesis and Integrin-mediated cell adhesion pathways, which were involved in cancer cell proliferation and metastasis, were showed to be significantly regulated in stages other than IA. CONCLUSION Genes involved in adipogenesis and Id pathways might play important roles in development of LADs. The similar trend of expression of the gene family members suggested coordinate regulation in tumor progression. Three pathways (Oxidative stress, Glycolysis/Gluconeogenesis and Integrin-mediated cell adhesion pathways) significantly regulated in stages other than stage IA suggested that genes and pathways conferring invasive character might be activated in the preinvasive stage IB, while the Oxidative stress and the Glycolysis/Gluconeogenesis pathways might have strong connections to cisplatin-based chemotherapy. The insignificantly regulated three pathways in stage IA might be used in early-stage detection of LAD.
Collapse
Affiliation(s)
- Chengwen Chen
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Zaravinos A, Lambrou GI, Volanis D, Delakas D, Spandidos DA. Spotlight on differentially expressed genes in urinary bladder cancer. PLoS One 2011; 6:e18255. [PMID: 21483670 PMCID: PMC3071699 DOI: 10.1371/journal.pone.0018255] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 03/01/2011] [Indexed: 01/30/2023] Open
Abstract
INTRODUCTION We previously identified common differentially expressed (DE) genes in bladder cancer (BC). In the present study we analyzed in depth, the expression of several groups of these DE genes. MATERIALS AND METHODS Samples from 30 human BCs and their adjacent normal tissues were analyzed by whole genome cDNA microarrays, qRT-PCR and Western blotting. Our attention was focused on cell-cycle control and DNA damage repair genes, genes related to apoptosis, signal transduction, angiogenesis, as well as cellular proliferation, invasion and metastasis. Four publicly available GEO Datasets were further analyzed, and the expression data of the genes of interest (GOIs) were compared to those of the present study. The relationship among the GOI was also investigated. GO and KEGG molecular pathway analysis was performed to identify possible enrichment of genes with specific biological themes. RESULTS Unsupervised cluster analysis of DNA microarray data revealed a clear distinction in BC vs. control samples and low vs. high grade tumors. Genes with at least 2-fold differential expression in BC vs. controls, as well as in non-muscle invasive vs. muscle invasive tumors and in low vs. high grade tumors, were identified and ranked. Specific attention was paid to the changes in osteopontin (OPN, SPP1) expression, due to its multiple biological functions. Similarly, genes exhibiting equal or low expression in BC vs. the controls were scored. Significant pair-wise correlations in gene expression were scored. GO analysis revealed the multi-facet character of the GOIs, since they participate in a variety of mechanisms, including cell proliferation, cell death, metabolism, cell shape, and cytoskeletal re-organization. KEGG analysis revealed that the most significant pathway was that of Bladder Cancer (p = 1.5×10(-31)). CONCLUSIONS The present work adds to the current knowledge on molecular signature identification of BC. Such works should progress in order to gain more insight into disease molecular mechanisms.
Collapse
Affiliation(s)
- Apostolos Zaravinos
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - George I. Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, University of Athens, Athens, Greece
| | - Dimitrios Volanis
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
- Department of Urology, Asklipieio General Hospital, Voula, Athens, Greece
| | - Dimitris Delakas
- Department of Urology, Asklipieio General Hospital, Voula, Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
- * E-mail:
| |
Collapse
|
17
|
Jahns F, Wilhelm A, Jablonowski N, Mothes H, Radeva M, Wölfert A, Greulich KO, Glei M. Butyrate suppresses mRNA increase of osteopontin and cyclooxygenase-2 in human colon tumor tissue. Carcinogenesis 2011; 32:913-20. [PMID: 21459756 DOI: 10.1093/carcin/bgr061] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The short chain fatty acid (SCFA) butyrate, a product of fermentation of dietary fiber in the human colon, is found to exert multiple regulatory processes in colon carcinogenesis. The aim of this study was to find out whether butyrate affects the tumor-promoting genes osteopontin (OPN) and cyclooxygenase (COX)-2, their respective proteins and/or their functional activity in matched normal, adenoma and tumor colon tissues obtained from 20 individuals at colon cancer surgery. Quantitative real-time polymerase chain reaction experiments showed increased levels of OPN and COX-2 messenger RNA in tumor tissues when compared with the adjacent normal samples (P < 0.001). The addition of butyrate reduced OPN and COX-2 mRNA expression in all tissue types compared with the related medium controls (tumor: P < 0.05). In tumor samples, a downregulation of up to median 35% (COX-2) and 50% (OPN) was observed, respectively. Thereby, tumors with lower levels of OPN basal expression were more sensitive to inhibition and vice versa for COX-2 in normal tissue. At the protein and enzyme level, which were determined by using western blot and enzyme immunometric assays, the impact of the SCFA was not clearly visible anymore. The active proteins of OPN and COX-2 (determined by prostaglandin E(2)) were found to correlate with their respective mRNA expression only in 50-63% of analyzed donors. For the first time, our data reveal new insights into the chemoprotective potential of butyrate by showing the suppression of OPN and COX-2 mRNA in primary human colon tissue with the strongest effects observed in tumors.
Collapse
Affiliation(s)
- F Jahns
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller-University Jena, Dornburger Straße 24, 07743 Jena, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
ING2 (inhibitor of growth 2) is a candidate tumor-suppressor gene involved in cell cycle control, apoptosis and senescence. Although the functions of ING2 within the chromatin remodeling complex Sin3A/histone deacetylase (HDAC) and in the p53 pathway have been described, how ING2 itself is regulated remains unknown. In this study we report for the first time that ING2 can be sumoylated by small ubiquitin-like modifier 1 (SUMO1) on lysine 195 both in vitro and in vivo. Strikingly, ING2 sumoylation enhances its association with Sin3a. We provide evidences that ING2 can bind to the promoter of genes to mediate their expression and that sumoylation of ING2 is required for this binding to some of these genes. Among them, we identified the gene TMEM71 (transmembrane protein 71), whose expression is regulated by ING2 sumoylation. ING2 must be sumoylated to bind to the promoter of TMEM71 and to recruit the Sin3A chromatin-modifying complex to this promoter, in order to regulate TMEM71 transcription. Hence, sumoylation of ING2 enhances its binding to the Sin3A/HDAC complex and is required to regulate gene transcriptions.
Collapse
|
19
|
Tavares TS, Nanus D, Yang XJ, Gudas LJ. Gene microarray analysis of human renal cell carcinoma: the effects of HDAC inhibition and retinoid treatment. Cancer Biol Ther 2008; 7:1607-18. [PMID: 18769122 PMCID: PMC3060607 DOI: 10.4161/cbt.7.10.6584] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitor treatments can augment the anti-tumor effects of retinoids in renal cancer cells. We studied the effects of the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) and 13-cis retinoic acid (cRA) on two human renal cell carcinoma (RCC) lines. Cells were cultured in the presence of each drug for six days to determine the responses to monotherapy and to combination therapy. The proliferation of SKRC06 was inhibited with cRA treatment; the proliferation of SKRC39 was not. However, both RCC lines were sensitive to growth inhibition by dibutyryl cyclic AMP, with or without 13-cis RA. SAHA alone also reduced cell proliferation in both cell lines. To identify the alterations in gene expression that correlate with the responsiveness to treatment, gene microarray analyses were performed. Several retinoid-regulated genes exhibited much higher mRNA levels in SKRC06 than in SKRC39, even in the absence of drugs; these included crabp2, rargamma and cyp26A1. Combination treatment of cells with both SAHA and cRA induced several transcripts with known anti-cancer/immunomodulatory effects, including dhrs9, gata3, il1beta, phlda1, txk and vhl. Immunostaining confirmed the decreased expression of gata3 in human RCC specimens compared to normal kidney. Together, our results show that treatment of RCC with cRA and/or SAHA increases the expression of several genes and gene families that result in reduced cell proliferation.
Collapse
Affiliation(s)
- Trisha S. Tavares
- Department of Pharmacology; Weill Cornell Medical College of Cornell University; New York, New York USA
| | - David Nanus
- Department of Medicine; Weill Cornell Medical College of Cornell University; New York, New York USA
| | - Ximing J. Yang
- Department of Pathology; Weill Cornell Medical College of Cornell University; New York, New York USA
| | - Lorraine J. Gudas
- Department of Pharmacology; Weill Cornell Medical College of Cornell University; New York, New York USA
- Department of Medicine; Weill Cornell Medical College of Cornell University; New York, New York USA
| |
Collapse
|
20
|
Song JY, Lee JK, Lee NW, Jung HH, Kim SH, Lee KW. Microarray analysis of normal cervix, carcinoma in situ, and invasive cervical cancer: identification of candidate genes in pathogenesis of invasion in cervical cancer. Int J Gynecol Cancer 2008; 18:1051-9. [PMID: 18217980 DOI: 10.1111/j.1525-1438.2007.01164.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The objective of this study was to identify genes that are related to pathogenesis of carcinoma in situ (CIS) to invasive cervical cancer with the use of oligonucleotide microarray and reverse transcription-polymerase chain reaction (RT-PCR). Each two cases of normal cervix, CIS, and invasive cervical cancer were investigated with DNA microarray technology. Differential gene expression profiles among them were analyzed. Expression levels of selected genes from the microarray results were confirmed by RT-PCR. The expressions of 15,286 genes were compared and 458 genes were upregulated or downregulated by twofold or more compared with each other group. Among 458 genes, 22 genes were upregulated and 40 genes were downregulated by twofold or more in invasive cervical cancer group compared with CIS group. RT-PCR analysis confirmed upregulation of 18 genes and downregulation of 5 genes in invasive cervical cancer group. RBP1, TFRC, SPP1, SAA1, ARHGAP8, and NDRG1, which were upregulated, and GATA3, PLAGL1, APOD, DUSP1, and CYR61, which were downregulated, were considered as candidate genes associated with invasion of cervical cancer.
Collapse
Affiliation(s)
- J Y Song
- Department of Obstetrics and Gynecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
21
|
Bean C, Facchinello N, Faulkner G, Lanfranchi G. The effects of Ankrd2 alteration indicate its involvement in cell cycle regulation during muscle differentiation. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1783:1023-35. [PMID: 18302940 DOI: 10.1016/j.bbamcr.2008.01.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 01/16/2008] [Accepted: 01/24/2008] [Indexed: 10/22/2022]
Abstract
Ankrd2 is a member of the Muscle Ankyrin Repeat Protein family (MARPs), consisting of sarcomere-associated proteins that can also localize in the nucleus. There are indications that MARPs might function as shuttle proteins between the cytoplasm and nucleus, likely sending information to the nucleus concerning the changes in the structure or function of the contractile machinery. Even though recent findings suggest that the MARP gene family is not essential for the basal functioning of skeletal muscle, its influence on the gene expression program of skeletal muscle cells was highlighted. To investigate this regulatory role we produced and examined both morphological and functional features of myocytes stable overexpressing or silencing the Ankrd2 protein. The transcriptional profiles of the myocytes revealed that the molecular pathways perturbed by changes in Ankrd2 protein level are congruent with the morpho-physiological and biochemical data obtained in Ankrd2-modified myoblasts induced to differentiate. Our results suggest that Ankrd2 gives an important contribution to the coordination of proliferation and apoptosis during myogenic differentiation in vitro, mainly through the p53 network.
Collapse
Affiliation(s)
- Camilla Bean
- Department of Biology and CRIBI Biotechnology Centre, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | | | | | | |
Collapse
|
22
|
Abstract
Osteopontin is a secreted phosphoprotein that has been implicated as an important mediator of tumor metastasis and has been investigated for use as a biomarker for advanced disease and as a potential therapeutic target in the regulation of cancer metastasis. The OPN DNA sequence is highly conserved and the protein contains several important functional domains including alpha(v)beta integrin and CD44 binding sites. High levels of OPN expression correlate with tumor invasion, progression or metastasis in multiple cancer. Studies demonstrate that osteopontin mediates the molecular mechanisms which determine metastatic spread, such as prevention of apoptosis, extracellular matrix proteolysis and remodeling, cell migration, evasion of host-immune cells and neovascularization. Transcriptional regulation of OPN is complex and involves multiple pathways, including AP-1, Myc, v-Src, Runx/CBF, TGF-B/BMPs/Smad/Hox, and Wnt/ss-catenin/APC/GSK-3ss/Tcf-4. The current state of knowledge of OPN biology suggests that it is an attractive target for therapeutic modulation of metastatic disease.
Collapse
|
23
|
Sasaki Y, Oshima Y, Koyama R, Maruyama R, Akashi H, Mita H, Toyota M, Shinomura Y, Imai K, Tokino T. Identification of flotillin-2, a major protein on lipid rafts, as a novel target of p53 family members. Mol Cancer Res 2008; 6:395-406. [PMID: 18296650 DOI: 10.1158/1541-7786.mcr-07-0108] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
p73 and p63 are members of the p53 gene family and have been shown to play an important role in development and homeostasis mainly by regulating the transcription of a variety of genes. A subset of these genes encodes secreted proteins and receptors that may be involved in the communication between adjacent cells. We report here that flotillin-2, a major hydrophobic protein on biomembrane microdomain lipid rafts, is a direct transcriptional target of the p53 family member genes. It has been suggested that such rafts could play an important role in many cellular processes including signal transduction, membrane trafficking, cytoskeletal organization, and pathogen entry. We found that the expression of flotillin-2 was specifically up-regulated by either TAp73beta or TAp63gamma, but not significantly by p53. In addition, flotillin-2 transcription is activated in response to cisplatin in a manner dependent on endogenous p73. By using small interference RNA designed to target p73, we showed that silencing endogenous p73 abolishes the induction of flotillin-2 transcription following cisplatin treatment. Furthermore, we identified a p73/p63-binding site located upstream of the flotillin-2 gene that is responsive to the p53 family members. This response element is highly conserved between humans and rodents. We also found that ectopic expression of TAp73 as well as TAp63 enhances signal transduction by assessing the interleukin-6-mediated phosphorylation of signal transducers and activators of transcription 3. Thus, in addition to direct transactivation, p53 family member genes enhance a set of cellular processes via lipid rafts.
Collapse
Affiliation(s)
- Yasushi Sasaki
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University, Chuo-ku, Sapporo, 060-8556 Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Guilarte TR, Burton NC, Verina T, Prabhu VV, Becker KG, Syversen T, Schneider JS. Increased APLP1 expression and neurodegeneration in the frontal cortex of manganese-exposed non-human primates. J Neurochem 2008; 105:1948-59. [PMID: 18284614 DOI: 10.1111/j.1471-4159.2008.05295.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Chronic manganese (Mn) exposure produces a neurological syndrome with psychiatric, cognitive, and parkinsonian features. Gene expression profiling in the frontal cortex of Cynomologous macaques receiving 3.3-5.0 mg Mn/kg weekly for 10 months showed that 61 genes were increased and four genes were decreased relative to controls from a total of 6766 genes. Gene changes were associated with cell cycle regulation, DNA repair, apoptosis, ubiquitin-proteasome system, protein folding, cholesterol homeostasis, axonal/vesicular transport, and inflammation. Amyloid-beta (Abeta) precursor-like protein 1, a member of the amyloid precursor protein family, was the most highly up-regulated gene. Immunohistochemistry confirmed increased amyloid precursor-like protein 1 protein expression and revealed the presence of diffuse Abeta plaques in Mn-exposed frontal cortex. Cortical neurons and white matter fibers from Mn-exposed animals accumulated silver grains indicative of on-going degeneration. Cortical neurons also exhibited nuclear hypertrophy, intracytoplasmic vacuoles, and apoptosis stigmata. p53 immunolabeling was increased in the cytoplasm of neurons and in the nucleus and processes of glial cells in Mn-exposed tissue. In summary, chronic Mn exposure produces a cellular stress response leading to neurodegenerative changes and diffuse Abeta plaques in the frontal cortex. These changes may explain the subtle cognitive deficits previously demonstrated in these same animals.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Chang PL, Harkins L, Hsieh YH, Hicks P, Sappayatosok K, Yodsanga S, Swasdison S, Chambers AF, Elmets CA, Ho KJ. Osteopontin expression in normal skin and non-melanoma skin tumors. J Histochem Cytochem 2007; 56:57-66. [PMID: 17938278 DOI: 10.1369/jhc.7a7325.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Osteopontin (OPN) is an adhesive, matricellular glycoprotein, whose expression is elevated in many types of cancer and has been shown to facilitate tumorigenesis in vivo. To understand the role of OPN in human skin cancer, this study is designed to determine whether OPN is expressed in premalignant [solar/actinic keratosis (AK)] and in malignant skin lesions such as squamous cell carcinomas (SCC) and basal cell carcinomas (BCC), as well as in normal skin exposed or not exposed to sunlight. Immunohistochemical analyses showed that OPN is expressed in SCC (20/20 cases) and in AK (16/16 cases), which are precursors to SCC, but is absent or minimally expressed in solid BCC (17 cases). However, positive staining for OPN was observed in those BCC that manifest differentiation toward epidermal appendages such as keratotic BCC. In sunlight-exposed normal skin, OPN is minimally expressed in the basal cell layer, but in contrast to those not exposed to sunlight, OPN is more prominent in the spinous cell layer with increasing intensity toward the granular cell layer. Additionally, OPN is expressed in the hair follicles, sebaceous glands, and sweat glands of normal skin. In conclusion, these data suggest that OPN is associated with keratinocyte differentiation and that it is expressed in AK and SCC, which have metastatic potential, but minimally expressed in solid BCC.
Collapse
Affiliation(s)
- Pi-Ling Chang
- Department of Nutrition Sciences, 311 Susan Mott Webb Nutrition Sciences Building, 1675 University Boulevard, University of Alabama at Birmingham, Birmingham, Alabama 35295-3360, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Graessmann M, Berg B, Fuchs B, Klein A, Graessmann A. Chemotherapy resistance of mouse WAP-SVT/t breast cancer cells is mediated by osteopontin, inhibiting apoptosis downstream of caspase-3. Oncogene 2006; 26:2840-50. [PMID: 17160024 DOI: 10.1038/sj.onc.1210096] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Impairment of the complex regulatory network of cell death and survival is frequently the reason for therapy resistance of breast cancer cells and a major cause of tumor progression. We established two independent cell lines from a fast growing mouse breast tumor (WAP-SVT/t transgenic animal). Cells from one line (ME-A cells) are sensitive to apoptotic stimuli such as growth factor depletion or treatment with antitumor agents (e.g. doxorubicin). Cells from the second line (ME-C cells), which carry a missense mutation at the p53 codon 242, are very insensitive to apoptotic stimuli. Co-cultivation experiments revealed that the ME-C cells mediate cell death resistance to the ME-A cells. Microarray and Western blot analysis showed that osteopontin (OPN) is selectively overexpressed by the ME-C cells. This glycoprotein is the most abundant protein secreted by the ME-C cells and we obtained strong indications that OPN is the main antiapoptotic factor. However, the OPN containing ME-C cell medium does not alter the expression level of pro- or antiapoptotic genes or known inhibitors of apoptosis (IAPs). Its signaling involves mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)1/2 as the kinase inhibitor PD98059 restores apoptosis but not the Akt inhibitor. In the ME-A cells, mitochondrial cytochrome c release occurs with and without external apoptotic stimuli. OPN containing ME-C cell medium does not prevent the mitochondrial cytochrome c release and caspase-9 processing. In serum starved ME-A cells, the OPN containing ME-C cell medium prevents caspase-3 activation. However, in doxorubicin-treated cells, although apoptosis is blocked, it does not inhibit caspase-3. This indicates that the ME-A cells distinguish between the initial apoptotic stimuli and that the cells possess a further uncharacterized control element acting downstream from caspase-3.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Antigens, Polyomavirus Transforming/genetics
- Apoptosis/genetics
- Caspase 3/physiology
- Culture Media, Serum-Free/pharmacology
- Cytochromes c/metabolism
- Doxorubicin/pharmacology
- Drug Resistance, Neoplasm/genetics
- JNK Mitogen-Activated Protein Kinases/metabolism
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Transgenic
- Milk Proteins/genetics
- Mitochondria/metabolism
- Models, Biological
- Osteopontin/metabolism
- Osteopontin/physiology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Graessmann
- Institut für Molekularbiologie und Bioinformatik, Charité Unversitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | |
Collapse
|
27
|
Khwaja FW, Svoboda P, Reed M, Pohl J, Pyrzynska B, Van Meir EG. Proteomic identification of the wt-p53-regulated tumor cell secretome. Oncogene 2006; 25:7650-61. [PMID: 17043663 DOI: 10.1038/sj.onc.1209969] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tumor-stroma interactions play a major role in tumor development, maintenance and progression. Yet little is known on how the genetic alterations that underlie cell transformation elicit cell extrinsic changes modulating heterotypic cell interactions. We hypothesized that these events involve a modification in the complement of secreted proteins by the cell, acting as mediators of intercellular communication. To test this hypothesis, we examined the role of wt-p53, a major tumor suppressor, on the tumor microenvironment through its regulation of secreted factors. Using a combination of 2-DE and cICAT proteomic techniques, we found a total of 111 secreted proteins, 39 of which showed enhanced and 21 inhibited secretion in response to wt-p53 expression. The majority of these were not direct targets of p53 transcription factor activity, suggesting a novel role for wt-p53 in the control of intracellular protein trafficking and/or secreted protein stability. Evidence for p53-controlled post-translational modifications on nine secreted proteins was also found. These findings will enhance our understanding of wt-p53 modulated interactions of the tumor with its environment.
Collapse
Affiliation(s)
- F W Khwaja
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
28
|
Celetti A, Testa D, Staibano S, Merolla F, Guarino V, Castellone MD, Iovine R, Mansueto G, Somma P, De Rosa G, Galli V, Melillo RM, Santoro M. Overexpression of the cytokine osteopontin identifies aggressive laryngeal squamous cell carcinomas and enhances carcinoma cell proliferation and invasiveness. Clin Cancer Res 2006; 11:8019-27. [PMID: 16299231 DOI: 10.1158/1078-0432.ccr-05-0641] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Osteopontin is a secreted cytokine that binds to the cell surface CD44v6 receptor. We studied osteopontin and CD44v6 expression in laryngeal squamous cell carcinomas and correlated osteopontin expression levels with clinicopathologic tumor features. EXPERIMENTAL DESIGN We used immunohistochemistry, immunoblotting, and reverse transcription-PCR to study osteopontin expression in 58 laryngeal squamous cell carcinomas. Cultured squamous carcinoma cells were treated with exogenous osteopontin or with RNA interference to knockdown osteopontin expression. RESULTS Osteopontin expression was higher in all the invasive carcinomas than in patient-matched normal mucosa. Its expression levels were significantly correlated with tumor stage and grade and with the presence of lymph node and distant metastases. Osteopontin positivity was negatively correlated with overall survival (P = 0.03). Osteopontin expression was paralleled by intense cell surface reactivity for CD44v6. Treatment of squamous carcinoma cells with recombinant osteopontin sharply increased proliferation and Matrigel invasion in comparison with the untreated cells parallel to activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/mitogen-activated protein kinase signaling cascade. Osteopontin knockdown by RNA interference, anti-CD44 antibodies, and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibition prevented these effects. CONCLUSIONS These results identify osteopontin as a marker and a potential therapeutic target in cases of aggressive laryngeal squamous cell carcinomas.
Collapse
Affiliation(s)
- Angela Celetti
- Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche/Dipartimento di Biologia e Patologia Cellulare e Molecolare, University of Naples Federico II, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Osteopontin (OPN) was initially isolated from bovine bone cortex, as a complex syalilated phospho-glyco-protein of around 60 kDa, with many postranslational modifications. It has been long considered a structural bone protein linking bone cells to the bone extracellular matrix (osteo : bone, pontin : bridge). It has been cloned for the first time in 1986. Since then, it was established that it is part of a protein family called SIBLINGs, which genes share common expression in bone and tooth, and encode among others a RGD motif. OPN is an intracellular as well as secreted protein, which binds to multiple organic or mineral ligands, like the integrin receptor alphaVbeta3, CD44, factor H and hydroxyapatite, depending on its final configuration (phosphorylation state). Pleiotropic functions of osteopontin have been demonstrated, and the osteopontin knock out phenotype in mice gave some new insight on the implication of the molecule in vivo. Osteopontin inhibits mineralization in bone and urine. Besides, it is a strong chemoattractive and proinflammatory molecule, implicated in tumors, like breast or prostate cancers, and in the defense against various infectious agents like tuberculosis, listeria or herpes. More recently, its key implication in TH1 mediated autoimmune diseases like multiple sclerosis and its animal model experimental autoimmune encephalomyelitis has been demonstrated. Osteopontin is a valuable therapeutic target in the animal model, and a biological tool correlating with clinical disease activity in humans. Structural, functional and pathological aspects of osteopontin are reviewed, as well as the osteopontin deficient phenotype in mouse.
Collapse
Affiliation(s)
- Dorothée Chabas
- Inserm U.546, Faculté de médecine Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
30
|
Chandar N, Swindle J, Szajkovics A, Kolman K. Relationship of bone morphogenetic protein expression during osteoblast differentiation to wild type p53. J Orthop Res 2005; 23:1345-53. [PMID: 15994055 DOI: 10.1016/j.orthres.2005.04.010.1100230616] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Accepted: 04/27/2005] [Indexed: 02/04/2023]
Abstract
We have previously shown p53 to have a specific role in osteoblast differentiation by its ability to regulate expression of certain bone specific proteins. In this study, we show mineralized matrix formation in vivo to be directly related to the presence of wild type p53 in osteoblastic osteosarcoma cells. In order to further understand the importance of p53 in differentiation, we investigated the relationship between p53 and Bone Morphogenetic Proteins (BMPs) (BMP 1, 2, 3A, 3B (GDF-10), 4, 5, 6, 7, 8A and 8B) during osteoblast differentiation. The expression of several BMPs were tested using RNase Protection Assay in differentiating ROS17/2.8 osteoblastic osteosarcoma cells. The expression of BMPs 1, 2, 3a, 3b and 7 showed time dependent modulation during in vitro differentiation. In order to determine if p53 has a role in this process, we used a murine osteosarcoma cell line stably expressing a temperature sensitive p53. Cells were exposed to ascorbic acid and glycerophosphates to hasten in vitro osteoblast differentiation and maintained either at 32 or 37 degrees C for expression of the wild type or mutant p53 phenotype. The expression of BMP-2, BMP-4 and BMP-7 were modulated in a p53 dependent fashion. We were able to confirm the p53 dependency of BMP-2 independently by RT-PCR. While BMP-2 expression was evident in the presence of both wild type and mutant p53, regulated expression was seen only in cells expressing wild type p53. Transient over expression of wild type p53 did not result in the same BMP-2 response as stable expression showing that the presence of p53 may be important for an orderly development of osteoblast differentiation rather than a direct effect on gene expression. The functional relationship between p53 and these bone specific markers is discussed.
Collapse
Affiliation(s)
- Nalini Chandar
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, 555, 31st Street, Downers Grove, IL 60515, USA.
| | | | | | | |
Collapse
|
31
|
di Pietro M, Sabates Bellver J, Menigatti M, Bannwart F, Schnider A, Russell A, Truninger K, Jiricny J, Marra G. Defective DNA mismatch repair determines a characteristic transcriptional profile in proximal colon cancers. Gastroenterology 2005; 129:1047-59. [PMID: 16143142 DOI: 10.1053/j.gastro.2005.06.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Accepted: 06/02/2005] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Colon cancers with defective DNA mismatch repair (MMR) have peculiar molecular, pathologic, and clinical features, including high-level microsatellite instability, conspicuous lymphocytic infiltration, preferential location in the proximal colon, and better prognosis. Our aim was to characterize the transcriptional profile of this colon cancer subset. METHODS An oligonucleotide microarray containing 12,625 probes was used to evaluate gene expression in 25 proximal colon cancers, 10 samples of normal colon mucosa, and 14 colon cancer cell lines. Transcriptional profiles of MMR-deficient cancers and cell lines were compared with those of their MMR-proficient counterparts. RESULTS Unsupervised analysis of microarray data showed that MMR status exerts a predominant influence on the gene expression profile of proximal colon cancers. Hierarchical clustering divided the cancers into 2 groups corresponding almost perfectly with their MMR status. Supervised analysis identified numerous gene expression changes that represent a genetic signature of MMR-deficient colon cancers. Changes in genes involved in apoptosis and the immune response were consistent with the better prognosis of MMR-deficient cancers. In MMR-deficient cancers and cell lines, 4-1BBL, a crucial gene in the anti-tumor immune response, was, respectively, 2.4 and 6.0 times more expressed than in their MMR-proficient counterparts. This difference was confirmed by quantitative reverse-transcription polymerase chain reaction and flow cytometric assessment of 4-1BBL protein expression in colon cancer cell lines. Our analysis also showed novel possible gene targets of microsatellite instability. CONCLUSIONS MMR inactivation produces distinct changes in the cellular messenger RNA pool, which is consistent with a unique tumorigenesis pathway.
Collapse
|
32
|
Kuznetsov NV, Andersson P, Gradin K, Stein PV, Dieckmann A, Pettersson S, Hanberg A, Poellinger L. The dioxin/aryl hydrocarbon receptor mediates downregulation of osteopontin gene expression in a mouse model of gastric tumourigenesis. Oncogene 2005; 24:3216-22. [PMID: 15735673 DOI: 10.1038/sj.onc.1208529] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The dioxin/aryl hydrocarbon receptor functions as a ligand-activated transcription factor regulating transcription of a battery of genes encoding primarily drug-metabolizing enzymes. Expression of a constitutively active mutant of the aryl hydrocarbon receptor (CA-AhR) in transgenic mice results in development of stomach tumours, correlating with increased mortality. We have used suppression subtractive hybridization techniques followed by macroarray analysis to elucidate which genes are differentially expressed during this process. In the glandular stomach of CA-AhR mice, we observed decreased mRNA expression of osteopontin (OPN), a noncollagenous protein of bone matrix that is also involved in several important functions including regulation of cytokine production, macrophage accumulation, cell motility and adhesion. Downregulated expression of OPN during tumour development was confirmed by RT-PCR and RNA blot analysis. Immunohistochemical analysis showed that this decrease was confined to the corpus region, correlating with the restricted localization of the tumours. Decreased OPN mRNA expression was also observed in other organs of CA-AhR mice. Taken together, these results show that OPN is negatively regulated by the dioxin receptor, and that downregulation of its expression correlates with development of stomach tumours in mice expressing a constitutively active mutant of dioxin receptor.
Collapse
|
33
|
Sasaki Y, Naishiro Y, Oshima Y, Imai K, Nakamura Y, Tokino T. Identification of pigment epithelium-derived factor as a direct target of the p53 family member genes. Oncogene 2005; 24:5131-6. [PMID: 15856012 DOI: 10.1038/sj.onc.1208695] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
p63 and p73 show a high degree of structural homology to p53 and are members of a family of transcriptional factors that can activate transcription of p53-responsive genes. p53 is mutated in more than 50% of human cancers, whereas p63 and p73 are rarely mutated. Studies of knockout mice also revealed an unexpected functional diversity among the p53 family. To determine how p63 and p73 are involved in tumorigenesis and normal development, we used cDNA microarray to examine 9216 genes in human colorectal cancer cells. We discovered that the expression of pigment epithelium-derived factor (PEDF) was specifically induced by either p63 or p73, but not by p53. We also report here that the PEDF gene contains a response element specific for p63 and p73 in its promoter region and is a direct target of p63 and p73. Collectively, p63 and p73 may be involved in cell fate by inducing PEDF expression.
Collapse
Affiliation(s)
- Yasushi Sasaki
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo 060-8556, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Sültmann H, Heydebreck AV, Huber W, Kuner R, Buneβ A, Vogt M, Gunawan B, Vingron M, Füzesí L, Poustka A. Gene Expression in Kidney Cancer Is Associated with Cytogenetic Abnormalities, Metastasis Formation, and Patient Survival. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.646.11.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Current diagnosis of renal cancer consists of histopathologic examination of tissue sections and classification into tumor stages and grades of malignancy. Until recently, molecular differences between tumor types were largely unknown. To examine such differences, we did gene expression measurements of 112 renal cell carcinoma and normal kidney samples on renal cell carcinoma–specific cDNA microarrays containing 4,207 genes and expressed sequence tags. The gene expression patterns showed deregulation of complete biological pathways in the tumors. Many of the molecular changes corresponded well to the histopathologic tumor types, and a set of 80 genes was sufficient to classify tumors with a very low error rate. Distinct gene expression signatures were associated with chromosomal abnormalities of tumor cells, metastasis formation, and patient survival. The data highlight the benefit of microarrays to detect novel tumor classes and to identify genes that are associated with patient variables and tumor properties.
Collapse
Affiliation(s)
- Holger Sültmann
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Anja von Heydebreck
- 2Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Berlin, Germany; and
| | - Wolfgang Huber
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Ruprecht Kuner
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Andreas Buneβ
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Markus Vogt
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Bastian Gunawan
- 3Institute of Pathology, University of Göttingen, Göttingen, Germany
| | - Martin Vingron
- 2Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Berlin, Germany; and
| | - Laszlo Füzesí
- 3Institute of Pathology, University of Göttingen, Göttingen, Germany
| | - Annemarie Poustka
- 1Division of Molecular Genome Analysis, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
35
|
Abstract
Osteopontin (OPN) is a glyco-phosphoprotein that is expressed and secreted by numerous human cancers. OPN functions in cell adhesion, chemotaxis, macrophage-directed interleukin-10 (IL-10) suppression, stress-dependent angiogenesis, prevention of apoptosis, and anchorage-independent growth of tumor cells by regulating cell-matrix interactions and cellular signaling through binding with integrin and CD44 receptors. While constitutive expression of OPN exists in several cell types, induced expression has been detected in T-lymphocytes, epidermal cells, bone cells, macrophages, and tumor cells in remodeling processes such as inflammation, ischemia-reperfusion, bone resorption, and tumor progression. Recently, substantial evidence has linked OPN with the regulation of metastatic spread by tumor cells. However, the molecular mechanisms that define the role of OPN in tumor metastasis are incompletely understood. Transcriptional regulators that contribute to the induction of OPN expression have received significant attention as potential modulators of the OPN-mediated metastatic phenotype. The following review will discuss the molecular structure of OPN, the evidence for its functional role in tumor cell metastasis, the downstream signals that activate invasive mechanisms, and the recent reports concerning regulation of OPN transcription.
Collapse
Affiliation(s)
- Philip Y Wai
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
36
|
Adachi K, Toyota M, Sasaki Y, Yamashita T, Ishida S, Ohe-Toyota M, Maruyama R, Hinoda Y, Saito T, Imai K, Kudo R, Tokino T. Identification of SCN3B as a novel p53-inducible proapoptotic gene. Oncogene 2004; 23:7791-8. [PMID: 15334053 DOI: 10.1038/sj.onc.1208067] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor suppressor p53 is a transcription factor that induces growth arrest and/or apoptosis in response to cellular stress. To identify novel p53-inducible genes, we compared the expression of genes in normal mouse embryo fibroblasts (MEFs) to p53-null cells by cDNA representational difference analysis. We report here that expression of endogenous sodium channel subunit beta 3 (SCN3B) is upregulated in mouse embryonic fibroblasts by DNA damage in a p53-dependent manner. In addition, we found that SCN3B levels are upregulated in human cancer cell lines by DNA damaging agents, as well as by overexpression of p53, but not significantly by p63 or p73. Furthermore, we identified two putative p53-binding sites upstream of the first exon (RE1) and in the third intron (RE2). The p53 protein can directly interact with the putative p53-binding sites in vivo, as assessed by chromatin immunoprecipitation. A reporter gene assay revealed that these two p53-binding sites are functional response elements. The SCN3B protein appears to be localized to the endoplasmic reticulum (ER). Introduction of the SCN3B gene into T98G and Saos2 cells potently suppressed colony formation. Furthermore, we found that adenovirus-mediated transfer of SCN3B induced apoptosis when combined with anticancer agents. The results presented here suggest that SCN3B mediates a p53-dependent apoptotic pathway and may be a candidate for gene therapy combined with anticancer drugs.
Collapse
Affiliation(s)
- Katsuya Adachi
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo 060-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Yoon KA, Nakamura Y, Arakawa H. Identification of ALDH4 as a p53-inducible gene and its protective role in cellular stresses. J Hum Genet 2004; 49:134-140. [PMID: 14986171 DOI: 10.1007/s10038-003-0122-3] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2003] [Accepted: 12/05/2003] [Indexed: 10/26/2022]
Abstract
To identify additional targets of p53, we used a cDNA microarray system to examine gene-expression patterns in response to enforced expression of exogenous p53 in p53-deficient cancer cells, and identified the aldehyde dehydrogenase 4 ( ALDH4) gene as a direct target of p53. ALDH4 is a mitochondrial-matrix NAD+-dependent enzyme catalyzing the second step of the proline degradation pathway. Expression of ALDH4 mRNA was induced in HCT116 cells in response to DNA damage caused by adriamycin treatment, in a p53-dependent manner. ALDH4 contains a potential p53 binding sequence in intron1 and the interaction of p53 with the site was shown by EMSA and ChIP assays. We confirmed p53-dependent transcriptional activity of the binding site by means of a reporter assay. Inhibition of ALDH4 expression by antisense oligonucleotides was able to enhance cell death induced by infection with Ad-p53. H1299 cells transformed to over-express ALDH4 showed significantly lower intracellular reactive oxygen species (ROS) levels than parental or control cells after treatment with hydrogen peroxide or UV. Those cells were also resistant to cell damage caused by hydrogen peroxide. These results suggest that p53 might play a protective role against cell damage induced by generation of intracellular ROS, through transcriptional activation of ALDH4.
Collapse
MESH Headings
- 1-Pyrroline-5-Carboxylate Dehydrogenase
- Aldehyde Dehydrogenase/genetics
- Aldehyde Dehydrogenase/physiology
- Antibiotics, Antineoplastic/pharmacology
- Apoptosis
- Binding Sites
- Blotting, Northern
- Cell Cycle
- Cell Line
- Cell Line, Tumor
- Cell Survival
- Chromatin/metabolism
- DNA Damage
- DNA, Complementary/metabolism
- Doxorubicin/pharmacology
- Flow Cytometry
- Genes, Reporter
- Humans
- Hydrogen Peroxide/pharmacology
- Luciferases/metabolism
- Mitochondria/metabolism
- Models, Biological
- Oligonucleotide Array Sequence Analysis
- Oligonucleotides, Antisense/chemistry
- Precipitin Tests
- RNA, Messenger/metabolism
- Reactive Oxygen Species
- Reverse Transcriptase Polymerase Chain Reaction
- Stress, Physiological
- Time Factors
- Transcription, Genetic
- Transcriptional Activation
- Tumor Suppressor Protein p53/metabolism
- Ultraviolet Rays
Collapse
Affiliation(s)
- Kyong-Ah Yoon
- Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yusuke Nakamura
- Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Hirofumi Arakawa
- Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
- Cancer Medicine and Biophysics Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
38
|
Abstract
BACKGROUND Leiomyosarcomas (LMS) are a common subtype of soft tissue sarcoma. The molecular causes of the disease remain unclear. METHODS In the current study, gene expression in LMS, leiomyomas, and normal myometrium was examined. RNA was prepared and gene expression was determined using microarray analysis arrays containing approximately 12,000 known genes and 48,000 expressed sequence tags (ESTs). RESULTS A number of genes were found to be differentially expressed in these sample sets, and six genes including cyclin-dependent kinase inhibitor 2A, diaphanous (Drosophila homolog) 3, doublecortin, calpain 6, interleukin-17B, and proteolipid 1 were found to be overexpressed in LMS compared with normal myometrium and 18 other tissues. Sets of genes were identified whose expression could be used to cluster samples with either LMS, leiomyomas, or normal myometrium. CONCLUSIONS The authors concluded that differences in gene expression can be detected between LMS and leiomyomas, normal myometrium, and other tissues, and that these changes in gene expression may yield clues with regard to the pathophysiology of leiomyosarcoma.
Collapse
Affiliation(s)
- Keith M Skubitz
- Department of Medicine, University of Minnesota Medical School and the Masonic Cancer Center, MMC 286, University Hospital, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
39
|
Watanabe G, Nishimori H, Irifune H, Sasaki Y, Ishida S, Zembutsu H, Tanaka T, Kawaguchi S, Wada T, Hata JI, Kusakabe M, Yoshida K, Nakamura Y, Tokino T. Induction of tenascin-C by tumor-specific EWS-ETS fusion genes. Genes Chromosomes Cancer 2003; 36:224-32. [PMID: 12557222 DOI: 10.1002/gcc.10153] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ewing sarcoma (ES) and peripheral primitive neuroectodermal tumors (PNETs) are associated with a chromosomal translocation resulting in a fusion of the amino-terminus of EWS with the DNA-binding domain of an ETS transcription factor (most commonly FLI1 or ERG). Although previous reports suggested that these chimera proteins would act as aberrant transcription factors, their downstream targets have not been fully elucidated. To identify downstream targets of these EWS-ETS fusion proteins, we introduced EWS-ETS fusion constructs into a human fibrosarcoma cell line, HT-1080, by retroviral transduction. Here we report that Tenascin-C (TNC) is induced to a significantly higher level in cells expressing EWS-ETSs than in cells expressing normal ETSs. Furthermore, through use of an antisense cDNA expression vector we show that expression of endogenous TNC mRNA and protein were reduced coordinately with attenuation of EWS-FLI1 fusion protein expression. A chromatin immunoprecipitation assay showed direct interaction between the TNC promoter and the EWS-FLI1 fusion protein in vivo. In addition, a luciferase reporter assay revealed that EWS-ETSs upregulated the TNC gene through four ETS binding sites in the TNC promoter. High levels of TNC expression were observed in a subset of ES cell lines (3 of 6) and primary tumors (4 of 6). Together with previous studies showing that TNC expression is involved in the invasive and malignant phenotype of several tumor types, our data suggest that the oncogenic effect of EWS-ETS may be mediated in part by upregulating of TNC expression.
Collapse
MESH Headings
- DNA, Antisense/pharmacology
- DNA, Complementary/pharmacology
- Down-Regulation/drug effects
- Fibrosarcoma/genetics
- Fibrosarcoma/metabolism
- Fibrosarcoma/pathology
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genetic Vectors/genetics
- Genetic Vectors/metabolism
- Genetic Vectors/physiology
- Humans
- Oligonucleotide Array Sequence Analysis
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/immunology
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/physiology
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/immunology
- Proto-Oncogene Protein c-fli-1
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- Proto-Oncogene Proteins c-ets
- RNA, Messenger/biosynthesis
- RNA-Binding Protein EWS/genetics
- RNA-Binding Protein EWS/metabolism
- RNA-Binding Protein EWS/physiology
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/pathology
- Tenascin/biosynthesis
- Tenascin/genetics
- Transcription Factors/genetics
- Transcription Factors/immunology
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcriptional Activation/genetics
- Transcriptional Activation/physiology
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Goichi Watanabe
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Nishimori H, Sasaki Y, Yoshida K, Irifune H, Zembutsu H, Tanaka T, Aoyama T, Hosaka T, Kawaguchi S, Wada T, Hata JI, Toguchida J, Nakamura Y, Tokino T. The Id2 gene is a novel target of transcriptional activation by EWS-ETS fusion proteins in Ewing family tumors. Oncogene 2002; 21:8302-9. [PMID: 12447693 DOI: 10.1038/sj.onc.1206025] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2002] [Revised: 08/08/2002] [Accepted: 10/04/2002] [Indexed: 11/09/2022]
Abstract
We report here that the Id2 (inhibitor of DNA binding 2) gene is a novel target of transcriptional activation by EWS-FLI1 and EWS-ERG, two fusion proteins that characterize Ewing family tumors (EFTs). To identify downstream targets of these EWS-ETS fusion proteins, we introduced EWS-ETS fusion constructs into a human fibrosarcoma cell line by retroviral transduction. cDNA microarray analysis revealed that Id2 expression was up-regulated by introducing the EWS-ETS fusion gene but not by the normal full-length ETS gene. An Id2 promoter-luciferase reporter assay showed that transactivation by EWS-ETS involves the minimal Id2 promoter and may function in cooperation with c-Myc within the full-length regulatory region. A chromatin immunoprecipitation assay revealed direct interaction between the Id2 promoter and EWS-FLI1 fusion protein in vivo. Significantly higher expression of Id2 and c-Myc was observed in all of the six EFT cell lines examined compared to six other sarcoma cell lines. Moreover, high levels of Id2 expression were also observed in five of the six primary tumors examined. Id2 is generally thought to affect the balance between cell differentiation and proliferation in development and is highly expressed in several cancer types. Considering these previous studies, our data suggest that the oncogenic effect of EWS-ETS may be mediated in part by up-regulating Id2 expression. doi:10.1038/sj.onc.1206025
Collapse
Affiliation(s)
- Hiroyuki Nishimori
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo, 060-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|