1
|
Huang G, Ouyang M, Xiao K, Zhou H, Zhong Z, Long S, Li Z, Zhang Y, Li L, Xiang S, Ding X. AP-2α decreases TMZ resistance of recurrent GBM by downregulating MGMT expression and improving DNA damage. Life Sci 2024; 357:123111. [PMID: 39369843 DOI: 10.1016/j.lfs.2024.123111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
AIMS The incidence of recurrent gliomas is high, exerting low survival rates and poor prognoses. Transcription factor AP-2α has been reported to regulate the progression of primary glioblastoma (GBM). However, the function of AP-2α in recurrent gliomas is largely unclear. METHODS The expression of AP-2α and O6-methylguanine DNA-methyltransferase (MGMT) was detected in recurrent glioma tissues and cell lines by Western blots, the regulation mechanisms between AP-2α/MGMT promoter and RA/AP-2α promoter were studied by luciferase reporter assays, EMSA, and chIP assays. The effects of AP-2α and TMZ/RA treatment on cell viability in vitro and in vivo were investigated by MTT assays, γH2AX staining, comet assays and intracranial injection. KEY FINDINGS AP-2α expression negatively correlates with the expression of MGMT in glioma samples. AP-2α could directly bind with the promoter of the MGMT gene, suppresses transcriptional levels of MGMT and downregulate MGMT expression in TMZ-resistant U87MG-R and T98G cells, but TMZ treatment decreases AP-2α expression and increases MGMT expression. The extended TMZ treatment and increased TMZ concentrations reversed these effects. Moreover, AP-2α overexpression combines with TMZ to decrease cell viability, concurrently with improved DNA damage marker γH2AX. Furthermore, retinoic acid (RA) activates RAR/RXR heterodimers, which bind to RA-responsive elements (RAREs) of the AP-2α promoter, and activates AP-2α expression in recurrent glioma cells. Finally, in intracranial relapsed glioma mouse model, both RA and TMZ could retard tumor development and prolong the mouse survival. SIGNIFICANCE AP-2α activation by gene overexpression or RA treatment reveals the suppressive effects on glioma relapse, providing a novel therapeutic strategy against malignant refractory gliomas.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Middle Aged
- Antineoplastic Agents, Alkylating/pharmacology
- Brain Neoplasms/drug therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/pathology
- Brain Neoplasms/metabolism
- Cell Line, Tumor
- DNA Damage/drug effects
- DNA Modification Methylases/metabolism
- DNA Modification Methylases/genetics
- DNA Repair Enzymes/genetics
- DNA Repair Enzymes/metabolism
- Down-Regulation/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Glioblastoma/drug therapy
- Glioblastoma/genetics
- Glioblastoma/pathology
- Glioblastoma/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/metabolism
- Promoter Regions, Genetic
- Temozolomide/pharmacology
- Transcription Factor AP-2/genetics
- Transcription Factor AP-2/metabolism
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guixiang Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Kai Xiao
- Department of Neurosurgery, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhe Zhong
- Department of Neurosurgery, Hunan Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan 410013, China
| | - Shengwen Long
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yiru Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Changsha 410081, China.
| | - Shuanglin Xiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
2
|
Engin AB, Engin A. Next-Cell Hypothesis: Mechanism of Obesity-Associated Carcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:727-766. [PMID: 39287871 DOI: 10.1007/978-3-031-63657-8_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Higher body fat content is related to a higher risk of mortality, and obesity-related cancer represents approximately 40% of all cancer patients diagnosed each year. Furthermore, epigenetic mechanisms are involved in cellular metabolic memory and can determine one's predisposition to being overweight. Low-grade chronic inflammation, a well-established characteristic of obesity, is a central component of tumor development and progression. Cancer-associated adipocytes (CAA), which enhance inflammation- and metastasis-related gene sets within the cancer microenvironment, have pro-tumoral effects. Adipose tissue is a major source of the exosomal micro ribonucleic acids (miRNAs), which modulate pathways involved in the development of obesity and obesity-related comorbidities. Owing to their composition of cargo, exosomes can activate receptors at the target cell or transfer molecules to the target cells and thereby change the phenotype of these cells. Exosomes that are released into the extracellular environment are internalized with their cargo by neighboring cells. The tumor-secreted exosomes promote organ-specific metastasis of tumor cells that normally lack the capacity to metastasize to a specific organ. Therefore, the communication between neighboring cells via exosomes is defined as the "next-cell hypothesis." The reciprocal interaction between the adipocyte and tumor cell is realized through the adipocyte-derived exosomal miRNAs and tumor cell-derived oncogenic miRNAs. The cargo molecules of adipocyte-derived exosomes are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. RNA-induced silencing regulates gene expression through various mechanisms. Destabilization of DICER enzyme, which catalyzes the conversion of primary miRNA (pri-miRNA) to precursor miRNA (pre-miRNA), is an important checkpoint in cancer development and progression. Interestingly, adipose tissue in obesity and tumors share similar pathogenic features, and the local hypoxia progress in both. While hypoxia in obesity leads to the adipocyte dysfunction and metabolic abnormalities, in obesity-related cancer cases, it is associated with worsened prognosis, increased metastatic potential, and resistance to chemotherapy. Notch-interleukin-1 (IL-1)-Leptin crosstalk outcome is referred to as "NILCO effect." In this chapter, obesity-related cancer development is discussed in the context of "next-cell hypothesis," miRNA biogenesis, and "NILCO effect."
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
3
|
Jin C, Luo Y, Liang Z, Li X, Kołat D, Zhao L, Xiong W. Crucial role of the transcription factors family activator protein 2 in cancer: current clue and views. J Transl Med 2023; 21:371. [PMID: 37291585 PMCID: PMC10249218 DOI: 10.1186/s12967-023-04189-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
The transcription factor family activator protein 2 (TFAP2) is vital for regulating both embryonic and oncogenic development. The TFAP2 family consists of five DNA-binding proteins, including TFAP2A, TFAP2B, TFAP2C, TFAP2D and TFAP2E. The importance of TFAP2 in tumor biology is becoming more widely recognized. While TFAP2D is not well studied, here, we mainly focus on the other four TFAP2 members. As a transcription factor, TFAP2 regulates the downstream targets directly by binding to their regulatory region. In addition, the regulation of downstream targets by epigenetic modification, posttranslational regulation, and interaction with noncoding RNA have also been identified. According to the pathways in which the downstream targets are involved in, the regulatory effects of TFAP2 on tumorigenesis are generally summarized as follows: stemness and EMT, interaction between TFAP2 and tumor microenvironment, cell cycle and DNA damage repair, ER- and ERBB2-related signaling pathway, ferroptosis and therapeutic response. Moreover, the factors that affect TFAP2 expression in oncogenesis are also summarized. Here, we review and discuss the most recent studies on TFAP2 and its effects on carcinogenesis and regulatory mechanisms.
Collapse
Affiliation(s)
- Chen Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxiao Luo
- University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Chinese Academy for Medical Sciences Oxford Institute, Oxford, UK
| | - Xi Li
- Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Damian Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Linyong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Weixi Xiong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Kołat D, Kałuzińska Ż, Bednarek AK, Płuciennik E. WWOX Loses the Ability to Regulate Oncogenic AP-2γ and Synergizes with Tumor Suppressor AP-2α in High-Grade Bladder Cancer. Cancers (Basel) 2021; 13:2957. [PMID: 34204827 PMCID: PMC8231628 DOI: 10.3390/cancers13122957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/03/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
The cytogenic locus of the WWOX gene overlaps with the second most active fragile site, FRA16D, which is present at a higher frequency in bladder cancer (BLCA) patients with smoking habit, a known risk factor of this tumor. Recently, we demonstrated the relevance of the role of WWOX in grade 2 BLCA in collaboration with two AP-2 transcription factors whose molecular actions supported or opposed pro-cancerous events, suggesting a distinct character. As further research is needed on higher grades, the aim of the present study was to examine WWOX-AP-2 functionality in grade 3 and 4 BLCA using equivalent in vitro methodology with additional transcriptome profiling of cellular variants. WWOX and AP-2α demonstrated similar anti-cancer functionality in most biological processes with subtle differences in MMP-2/9 regulation; this contradicted that of AP-2γ, whose actions potentiated cancer progression. Simultaneous overexpression of WWOX and AP-2α/AP-2γ revealed that single discrepancies appear in WWOX-AP-2α collaboration but only at the highest BLCA grade; WWOX-AP-2α collaboration was considered anti-cancer. However, WWOX only appeared to have residual activity against oncogenic AP-2γ in grade 3 and 4: variants with either AP-2γ overexpression alone or combined WWOX and AP-2γ overexpression demonstrated similar pro-tumoral behavior. Transcriptome profiling with further gene ontology certified biological processes investigated in vitro and indicated groups of genes consisting of AP-2 targets and molecules worth investigation as biomarkers. In conclusion, tumor suppressor synergism between WWOX and AP-2α is unimpaired in high-grade BLCA compared to intermediate grade, yet the ability of WWOX to guide oncogenic AP-2γ is almost completely lost.
Collapse
Affiliation(s)
- Damian Kołat
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752 Lodz, Poland; (Ż.K.); (A.K.B.); (E.P.)
| | | | | | | |
Collapse
|
5
|
Kołat D, Kałuzińska Ż, Orzechowska M, Bednarek AK, Płuciennik E. Functional genomics of AP-2α and AP-2γ in cancers: in silico study. BMC Med Genomics 2020; 13:174. [PMID: 33213447 PMCID: PMC7678100 DOI: 10.1186/s12920-020-00823-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Among all causes of death, cancer is the most prevalent and is only outpaced by cardiovascular diseases. Molecular theory of carcinogenesis states that apoptosis and proliferation are regulated by groups of tumor suppressors or oncogenes. Transcription factors are example of proteins comprising representatives of both cancer-related groups. Exemplary family of transcription factors which exhibits dualism of function is Activating enhancer-binding Protein 2 (AP-2). Scientific reports concerning their function in carcinogenesis depend on particular family member and/or tumor type which proves the issue to be unsolved. Therefore, the present study examines role of the best-described AP-2 representatives, AP-2α and AP-2γ, through ontological analysis of their target genes and investigation what processes are differentially regulated in 21 cancers using samples deposited in Genomic Data Analysis Center (GDAC) Firehose. METHODS Expression data with clinical annotation was collected from TCGA-dedicated repository GDAC Firehose. Transcription factor targets were obtained from Gene Transcription Regulation Database (GTRD), TRANScription FACtor database (TRANSFAC) and Transcriptional Regulatory Relationships Unraveled by Sentence-based Text mining (TRRUST). Monocle3 R package was used for global samples profiling while Protein ANalysis THrough Evolutionary Relationships (PANTHER) tool was used to perform gene ontology analysis. RESULTS With RNA-seq data and Monocle3 or PANTHER tools we outlined differences in many processes and signaling pathways, separating tumor from normal tissues or tumors from each other. Unexpectedly, a number of alterations in basal-like breast cancer were identified that distinguished it from other subtypes, which could bring future clinical benefits. CONCLUSIONS Our findings indicate that while the AP-2α/γ role remains ambiguous, their activity is based on processes that underlie the cancer hallmarks and their expression could have potential in diagnosis of selected tumors.
Collapse
Affiliation(s)
- Damian Kołat
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland.
| | - Żaneta Kałuzińska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Magdalena Orzechowska
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, 90-752, Lodz, Poland
| |
Collapse
|
6
|
Bang J, Zippin JH. Cyclic adenosine monophosphate (cAMP) signaling in melanocyte pigmentation and melanomagenesis. Pigment Cell Melanoma Res 2020; 34:28-43. [PMID: 32777162 DOI: 10.1111/pcmr.12920] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
The second messenger cyclic adenosine monophosphate (cAMP) regulates numerous functions in both benign melanocytes and melanoma cells. cAMP is generated from two distinct sources, transmembrane and soluble adenylyl cyclases (tmAC and sAC, respectively), and is degraded by a family of proteins called phosphodiesterases (PDEs). cAMP signaling can be regulated in many different ways and can lead to varied effects in melanocytes. It was recently revealed that distinct cAMP signaling pathways regulate pigmentation by either altering pigment gene expression or the pH of melanosomes. In the context of melanoma, many studies report seemingly contradictory roles for cAMP in tumorigenesis. For example, cAMP signaling has been implicated in both cancer promotion and suppression, as well as both therapy resistance and sensitization. This conundrum in the field may be explained by the fact that cAMP signals in discrete microdomains and each microdomain can mediate differential cellular functions. Here, we review the role of cAMP signaling microdomains in benign melanocyte biology, focusing on pigmentation, and in melanomagenesis.
Collapse
Affiliation(s)
- Jakyung Bang
- Department of Dermatology, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Jonathan H Zippin
- Department of Dermatology, Joan and Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
7
|
Huang W, Zhong Z, Luo C, Xiao Y, Li L, Zhang X, Yang L, Xiao K, Ning Y, Chen L, Liu Q, Hu X, Zhang J, Ding X, Xiang S. The miR-26a/AP-2α/Nanog signaling axis mediates stem cell self-renewal and temozolomide resistance in glioma. Am J Cancer Res 2019; 9:5497-5516. [PMID: 31534499 PMCID: PMC6735392 DOI: 10.7150/thno.33800] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
Aberrant expression of transcription factor AP-2α has been functionally associated with various cancers, but its clinical significance and molecular mechanisms in human glioma are largely elusive. Methods: AP-2α expression was analyzed in human glioma tissues by immunohistochemistry (IHC) and in glioma cell lines by Western blot. The effects of AP-2α on glioma cell proliferation, migration, invasion and tumor formation were evaluated by the 3-(4,5-dimethyNCthiazol-2-yl)-25-diphenyltetrazolium bromide (MTT) and transwell assays in vitro and in nude mouse models in vivo. The influence of AP-2α on glioma cell stemness was analyzed by sphere-formation, self-renewal and limiting dilution assays in vitro and in intracranial mouse models in vivo. The effects of AP-2α on temozolomide (TMZ) resistance were detected by the MTT assay, cell apoptosis, real-time PCR analysis, western blotting and mouse experiments. The correlation between AP-2α expression and the expression of miR-26a, Nanog was determined by luciferase reporter assays, electrophoretic mobility shift assay (EMSA) and expression analysis. Results: AP-2α expression was downregulated in 58.5% of glioma tissues and in 4 glioma cell lines. AP-2α overexpression not only reduced the proliferation, migration and invasion of glioma cell lines but also suppressed the sphere-formation and self-renewal abilities of glioma stem cells in vitro. Moreover, AP-2α overexpression inhibited subcutaneous and intracranial xenograft tumor growth in vivo. Furthermore, AP-2α enhanced the sensitivity of glioma cells to TMZ. Finally, AP-2α directly bound to the regulatory region of the Nanog gene, reduced Nanog, Sox2 and CD133 expression. Meanwhile, AP-2α indirectly downregulated Nanog expression by inhibiting the interleukin 6/janus kinase 2/signal transducer and activator of transcription 3 (IL6/JAK2/STAT3) signaling pathway, consequently decreasing O6-methylguanine methyltransferase (MGMT) and programmed death-ligand 1 (PD-L1) expression. In addition, miR-26a decreased AP-2α expression by binding to the 3' untranslated region (UTR) of AP-2α and reversed the tumor suppressive role of AP-2α in glioma, which was rescued by a miR-26a inhibitor. TMZ and the miR-26a inhibitor synergistically suppressed intracranial GSC growth. Conclusion: These results suggest that AP-2α reduces the stemness and TMZ resistance of glioma by inhibiting the Nanog/Sox2/CD133 axis and IL6/STAT3 signaling pathways. Therefore, AP-2α and miR-26a inhibition might represent a new target for developing new therapeutic strategies in TMZ resistance and recurrent glioma patients.
Collapse
|
8
|
Chachami G, Stankovic-Valentin N, Karagiota A, Basagianni A, Plessmann U, Urlaub H, Melchior F, Simos G. Hypoxia-induced Changes in SUMO Conjugation Affect Transcriptional Regulation Under Low Oxygen. Mol Cell Proteomics 2019; 18:1197-1209. [PMID: 30926672 PMCID: PMC6553927 DOI: 10.1074/mcp.ra119.001401] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
Hypoxia occurs in pathological conditions, such as cancer, as a result of the imbalance between oxygen supply and consumption by proliferating cells. HIFs are critical molecular mediators of the physiological response to hypoxia but also regulate multiple steps of carcinogenesis including tumor progression and metastasis. Recent data support that sumoylation, the covalent attachment of the Small Ubiquitin-related MOdifier (SUMO) to proteins, is involved in the activation of the hypoxic response and the ensuing signaling cascade. To gain insights into differences of the SUMO1 and SUMO2/3 proteome of HeLa cells under normoxia and cells grown for 48 h under hypoxic conditions, we employed endogenous SUMO-immunoprecipitation in combination with quantitative mass spectrometry (SILAC). The group of proteins whose abundance was increased both in the total proteome and in the SUMO IPs from hypoxic conditions was enriched in enzymes linked to the hypoxic response. In contrast, proteins whose SUMOylation status changed without concomitant change in abundance were predominantly transcriptions factors or transcription regulators. Particularly interesting was transcription factor TFAP2A (Activating enhancer binding Protein 2 alpha), whose sumoylation decreased on hypoxia. TFAP2A is known to interact with HIF-1 and we provide evidence that deSUMOylation of TFAP2A enhances the transcriptional activity of HIF-1 under hypoxic conditions. Overall, these results support the notion that SUMO-regulated signaling pathways contribute at many distinct levels to the cellular response to low oxygen.
Collapse
Affiliation(s)
- Georgia Chachami
- From the ‡Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
- ‡‡Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
| | - Nicolas Stankovic-Valentin
- §Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
| | - Angeliki Karagiota
- From the ‡Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
| | - Angeliki Basagianni
- From the ‡Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
| | - Uwe Plessmann
- ¶Bioanalytical Mass Spectrometry Group Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Henning Urlaub
- ¶Bioanalytical Mass Spectrometry Group Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
- ‖Bioanalytics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Frauke Melchior
- §Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
| | - George Simos
- From the ‡Laboratory of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
- **Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
9
|
Kołat D, Kałuzińska Ż, Bednarek AK, Płuciennik E. The biological characteristics of transcription factors AP-2α and AP-2γ and their importance in various types of cancers. Biosci Rep 2019; 39:BSR20181928. [PMID: 30824562 PMCID: PMC6418405 DOI: 10.1042/bsr20181928] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The Activator Protein 2 (AP-2) transcription factor (TF) family is vital for the regulation of gene expression during early development as well as carcinogenesis process. The review focusses on the AP-2α and AP-2γ proteins and their dualistic regulation of gene expression in the process of carcinogenesis. Both AP-2α and AP-2γ influence a wide range of physiological or pathological processes by regulating different pathways and interacting with diverse molecules, i.e. other proteins, long non-coding RNAs (lncRNA) or miRNAs. This review summarizes the newest information about the biology of two, AP-2α and AP-2γ, TFs in the carcinogenesis process. We emphasize that these two proteins could have either oncogenic or suppressive characteristics depending on the type of cancer tissue or their interaction with specific molecules. They have also been found to contribute to resistance and sensitivity to chemotherapy in oncological patients. A better understanding of molecular network of AP-2 factors and other molecules may clarify the atypical molecular mechanisms occurring during carcinogenesis, and may assist in the recognition of new diagnostic biomarkers.
Collapse
Affiliation(s)
- Damian Kołat
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Żaneta Kałuzińska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
10
|
The DCBLD receptor family: emerging signaling roles in development, homeostasis and disease. Biochem J 2019; 476:931-950. [PMID: 30902898 DOI: 10.1042/bcj20190022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/20/2019] [Accepted: 03/04/2019] [Indexed: 02/08/2023]
Abstract
The discoidin, CUB, and LCCL domain-containing (DCBLD) receptor family are composed of the type-I transmembrane proteins DCBLD1 and DCBLD2 (also ESDN and CLCP1). These proteins are highly conserved across vertebrates and possess similar domain structure to that of neuropilins, which act as critical co-receptors in developmental processes. Although DCBLD1 remains largely uncharacterized, the functional and mechanistic roles of DCBLD2 are emerging. This review provides a comprehensive discussion of this presumed receptor family, ranging from structural and signaling aspects to their associations with cancer, physiology, and development.
Collapse
|
11
|
Pavone V, Chisari E, Vescio A, Lizzio C, Sessa G, Testa G. Aetiology of Legg-Calvé-Perthes disease: A systematic review. World J Orthop 2019; 10:145-165. [PMID: 30918798 PMCID: PMC6429000 DOI: 10.5312/wjo.v10.i3.145] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/06/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Legg-Calvé-Perthes disease (LCPD) is a clinical condition affecting the femoral head of children during their growth. Its prevalence is set to be between 0.4/100000 to 29.0/100000 children less than 15 years of age with a peak of incidence in children aged from 4 years to 8 years. LCPD aetiology has been widely studied, but it is still poorly understood. AIM To analyse the available literature to document the up-to-date evidence on LCPD aetiology. METHODS A systematic review of the literature was performed regarding LCPD aetiology, using the following inclusion criteria: studies of any level of evidence, reporting clinical or preclinical results and dealing with the aetiology or pathogenesis of LCPD. Two reviewers searched the PubMed and Science Direct databases from their date of inception to the 20th of May 2018 in accordance with the Preferred Reporting Items for Systemic Reviews and Meta-Analyses guidelines. To achieve the maximum sensitivity of the search strategy, we combined the terms: ''Perthes disease OR LCPD OR children avascular femoral head necrosis" with "pathology OR aetiology OR biomechanics OR genetics" as either key words or MeSH terms. RESULTS We include 64 articles in this review. The available evidence on LCPD aetiology is still debated. Several hypotheses have been researched, but none of them was found decisive. While emerging evidence showed the role of environmental risk factors and evidence from twin studies did not support a major role for genetic factors, a congenital or acquired predisposition cannot be excluded in disease pathogenesis. One of the most supported theories involved mechanical induced ischemia that evolved into avascular necrosis of the femoral head in sensible patients. CONCLUSION The literature available on the aetiology of LCPD presents major limitations in terms of great heterogeneity and a lack of high-profile studies. Although a lot of studies focused on the genetic, biomechanical and radiological background of the disease, there is a lack of consensus on one or multiple major actors of the etiopathogenesis. More studies are needed to understand the complex and multifactorial genesis of the avascular necrosis characterizing the disease.
Collapse
Affiliation(s)
- Vito Pavone
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| | - Emanuele Chisari
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| | - Andrea Vescio
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| | - Claudio Lizzio
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| | - Giuseppe Sessa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| | - Gianluca Testa
- Department of General Surgery and Medical Surgical Specialties, Section of Orthopaedics and Traumatology, University Hospital Policlinico-Vittorio Emanuele, University of Catania, Catania 95100, Italy
| |
Collapse
|
12
|
Su F, Zhang W, Zhang D, Zhang Y, Pang C, Huang Y, Wang M, Cui L, He L, Zhang J, Zou L, Zhang J, Li W, Li L, Shao J, Ma J, Xiao F, Liu M. Spatial Intratumor Genomic Heterogeneity within Localized Prostate Cancer Revealed by Single-nucleus Sequencing. Eur Urol 2018; 74:551-559. [DOI: 10.1016/j.eururo.2018.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022]
|
13
|
Liu Y, Yang Y, Wang T, Wang L, Wang X, Li T, Shi Y, Wang Y. Long non-coding RNA CCAL promotes hepatocellular carcinoma progression by regulating AP-2α and Wnt/β-catenin pathway. Int J Biol Macromol 2017; 109:424-434. [PMID: 29275200 DOI: 10.1016/j.ijbiomac.2017.12.110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/26/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Long non-coding RNAs are emerging as key molecules in cancer progression. LncRNA-CCAL has shown to be highly expressed and important in regulating CRC and osteosarcoma development. Nevertheless, the expression and mechanism of CCAL in HCC is still not well understood. METHODS qRT-PCR and ISH were used to evaluate CCAL expression in HCC tissues and cell lines. Histone H3 methylation and acetylation levels across CCAL promoter region were examined by chromatin immunoprecipitation assays. Transfection of Lv-CCAL-shRNAs into HCC cell lines was used to evaluate cellular invasion and proliferation. The influence of CCAL depletion on AP-2α expression and Wnt/β-catenin pathway was analyzed by qRT-PCR, western blot and immunofluorescence. RESULTS Higher expression of CCAL was found in HCC tumor tissues compared with normal tissues, and was associated with tumor metastasis and TNM stage. Furthermore, the decreased histone H3 methylation and increased histone H3 acetylation across CCAL promoter region contributed to the upregulation of CCAL in HCC. Moreover, the depletion of CCAL inhibited HCC cellular invasion and proliferation, and promoted cell apoptosis. In addition, CCAL depletion up-regulated AP-2α expression and inhibited Wnt/β-catenin pathway activation. CONCLUSIONS CCAL has an important role in hepatic carcinogenesis and may serve as a new target for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Yu Liu
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Ye Yang
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Tianlong Wang
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Li Wang
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Xin Wang
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Tiemin Li
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Yue Shi
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China
| | - Yawei Wang
- Department of geriatric surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, PR China.
| |
Collapse
|
14
|
De Luca A, Carpanese D, Rapanotti MC, Viguria TMS, Forgione MA, Rotili D, Fulci C, Iorio E, Quintieri L, Chimenti S, Bianchi L, Rosato A, Caccuri AM. The nitrobenzoxadiazole derivative MC3181 blocks melanoma invasion and metastasis. Oncotarget 2017; 8:15520-15538. [PMID: 28107182 PMCID: PMC5362503 DOI: 10.18632/oncotarget.14690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/27/2016] [Indexed: 02/01/2023] Open
Abstract
The novel nitrobenzoxadiazole (NBD) derivative MC3181 is endowed with remarkable therapeutic activity in mice bearing both sensitive and vemurafenib-resistant human melanoma xenografts. Here, we report that subtoxic concentrations of this compound significantly reduced invasiveness of BRAF-V600D mutated WM115 and WM266.4 melanoma cell lines derived from the primary lesion and related skin metastasis of the same patient, respectively. The strong antimetastatic activity of MC3181 was observed in both 2D monolayer cultures and 3D multicellular tumor spheroids, and confirmed in vivo by the significant decrease in the number of B16-F10 melanoma lung metastases in drug-treated mice. Our data also show that MC3181 affects the lactate production in the high glycolytic WM266.4 cell line. To unveil the MC3181 mechanism of action, we analyzed the ability of MC3181 to affect the degree of activation of different MAPK pathways, as well as the expression/activity levels of several proteins involved in angiogenesis, invasion, and survival (i.e. AP2, MCAM/MUC18, N-cadherin, VEGF and MMP-2). Our data disclosed both a decrease of the phospho-active form of JNK and an increased expression of the transcription factor AP2, events that occur in the very early phase of drug treatment and may be responsible of the antimetastatic effects of MC3181.
Collapse
Affiliation(s)
- Anastasia De Luca
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Debora Carpanese
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | | | | | | | - Dante Rotili
- Department of Drug Chemistry and Technologies, "Sapienza" University, 00185 Rome, Italy
| | - Chiara Fulci
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Egidio Iorio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Sergio Chimenti
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Luca Bianchi
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy.,Istituto Oncologico Veneto IOV-IRCCS, 35128 Padova, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
15
|
Dhingra P, Martinez-Fundichely A, Berger A, Huang FW, Forbes AN, Liu EM, Liu D, Sboner A, Tamayo P, Rickman DS, Rubin MA, Khurana E. Identification of novel prostate cancer drivers using RegNetDriver: a framework for integration of genetic and epigenetic alterations with tissue-specific regulatory network. Genome Biol 2017; 18:141. [PMID: 28750683 PMCID: PMC5530464 DOI: 10.1186/s13059-017-1266-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 06/27/2017] [Indexed: 11/22/2022] Open
Abstract
We report a novel computational method, RegNetDriver, to identify tumorigenic drivers using the combined effects of coding and non-coding single nucleotide variants, structural variants, and DNA methylation changes in the DNase I hypersensitivity based regulatory network. Integration of multi-omics data from 521 prostate tumor samples indicated a stronger regulatory impact of structural variants, as they affect more transcription factor hubs in the tissue-specific network. Moreover, crosstalk between transcription factor hub expression modulated by structural variants and methylation levels likely leads to the differential expression of target genes. We report known prostate tumor regulatory drivers and nominate novel transcription factors (ERF, CREB3L1, and POU2F2), which are supported by functional validation.
Collapse
Affiliation(s)
- Priyanka Dhingra
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA
| | - Alexander Martinez-Fundichely
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA
| | - Adeline Berger
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, 10065, USA
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
- Cancer Program, The Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, 02142, USA
| | - Andre Neil Forbes
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA
| | - Eric Minwei Liu
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA
| | - Deli Liu
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA
- Department of Urology, Weill Cornell Medical College, New York, New York, 10065, USA
| | - Andrea Sboner
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA
| | - Pablo Tamayo
- Cancer Program, The Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, 02142, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, 10065, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York, 10065, USA.
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York, 10065, USA
| | - Ekta Khurana
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, 10065, USA.
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York, 10021, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital-Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Weill Cornell Medical College, New York, New York, 10065, USA.
| |
Collapse
|
16
|
Wang F, Huang W, Hu X, Chen C, Li X, Qiu J, Liang Z, Zhang J, Li L, Wang X, Ding X, Xiang S, Zhang J. Transcription factor AP-2β suppresses cervical cancer cell proliferation by promoting the degradation of its interaction partner β-catenin. Mol Carcinog 2017; 56:1909-1923. [PMID: 28277615 DOI: 10.1002/mc.22646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 12/21/2022]
Abstract
Transcription factor AP-2β mediates the transcription of a number of genes implicated in mammalian development, cell proliferation, and carcinogenesis. Although the expression pattern of AP-2β has been analyzed in cervical cancer cell lines, the functions and molecular mechanism of AP-2β are unknown. Here, we found that AP-2β significantly inhibits TCF/LEF reporter activity. Moreover, AP-2β and β-catenin interact both in vitro through GST pull-down assays and in vivo by co-immunoprecipitation. We further identified the interaction regions to the DNA-binding domain of AP-2β and the 1-9 Armadillo repeats of β-catenin. Moreover, AP-2β binds with β-TrCP and promotes the degradation of endogenous β-catenin via the proteasomal degradation pathway. Immunohistochemistry analysis revealed a negative correlation between the two proteins in cervical cancer tissues and cell lines. Finally, functional analysis showed that AP-2β suppresses cervical cancer cell growth in vitro and in vivo by inhibiting the expression of Wnt downstream genes. Taken together, these findings demonstrated that AP-2β functions as a novel inhibitor of the Wnt/β-catenin signaling pathway in cervical cancer.
Collapse
Affiliation(s)
- Fangmei Wang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Wenhuan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Cheng Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Xinxin Li
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Junlu Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Zhongheng Liang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Jianmei Zhang
- Reproductive Medicine Center, Changsha Hospital for Maternal & Child Health Care, Changsha, Hunan, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoqing Wang
- Xiangya Second Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Changsha, Hunan, China
| |
Collapse
|
17
|
Ge L, Vujanovic NL. Soluble TNF Regulates TACE via AP-2α Transcription Factor in Mouse Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2016; 198:417-427. [PMID: 27852742 DOI: 10.4049/jimmunol.1600524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 10/25/2016] [Indexed: 01/12/2023]
Abstract
Dendritic cells (DCs), the essential immunoregulatory and APCs, are major producers of the central mediator of inflammation, soluble TNF-α (sTNF). sTNF is generated by TNF-α converting enzyme (TACE) proteolytic release of the transmembrane TNF (tmTNF) ectodomain. The mechanisms of TACE and sTNF regulation in DCs remain elusive. This study newly defines that sTNF regulates TACE in mouse DCs by engaging the AP-2α transcription factor. We found that the expression of AP-2α was higher, whereas the expression and activity of TACE were lower, in wild-type DCs (wtDCs) than in TNF knockout (TNFko) DCs. Exogenous sTNF rapidly and simultaneously induced increases of AP-2α expression and decreases of TACE expression and activity in wtDCs and TNFko DCs, indicating that AP-2α and TACE are inversely dependent on sTNF and are functionally associated. To define this functional association, we identified an AP-2α binding site in TACE promoter and demonstrated, using EMSAs and chromatin immunoprecipitation assays, that AP-2α could bind to TACE promoter in a TNF-dependent manner. Additionally, sTNF simultaneously enhanced AP-2α expression and decreased TACE promoter luciferase activity in DCs. Similarly, transfection of AP-2α cDNA decreased TACE promoter luciferase activity, TACE expression, and TACE enzymatic activity in wtDCs or TNFko DCs. In contrast, transfection of AP-2α small interfering RNA increased TACE promoter luciferase activity, TACE expression, and TACE enzymatic activity in wtDCs. These results show that TACE is a target of, and is downregulated by, sTNF-induced AP-2α transcription factor in DCs.
Collapse
Affiliation(s)
- Lisheng Ge
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| | - Nikola L Vujanovic
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232; .,Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232; and.,VA Pittsburgh Healthcare System, Pittsburgh, PA 15261
| |
Collapse
|
18
|
HIPSTR and thousands of lncRNAs are heterogeneously expressed in human embryos, primordial germ cells and stable cell lines. Sci Rep 2016; 6:32753. [PMID: 27605307 PMCID: PMC5015059 DOI: 10.1038/srep32753] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/11/2016] [Indexed: 01/02/2023] Open
Abstract
Eukaryotic genomes are transcribed into numerous regulatory long non-coding RNAs (lncRNAs). Compared to mRNAs, lncRNAs display higher developmental stage-, tissue-, and cell-subtype-specificity of expression, and are generally less abundant in a population of cells. Despite the progress in single-cell-focused research, the origins of low population-level expression of lncRNAs in homogeneous populations of cells are poorly understood. Here, we identify HIPSTR (Heterogeneously expressed from the Intronic Plus Strand of the TFAP2A-locus RNA), a novel lncRNA gene in the developmentally regulated TFAP2A locus. HIPSTR has evolutionarily conserved expression patterns, its promoter is most active in undifferentiated cells, and depletion of HIPSTR in HEK293 and in pluripotent H1BP cells predominantly affects the genes involved in early organismal development and cell differentiation. Most importantly, we find that HIPSTR is specifically induced and heterogeneously expressed in the 8-cell-stage human embryos during the major wave of embryonic genome activation. We systematically explore the phenomenon of cell-to-cell variation of gene expression and link it to low population-level expression of lncRNAs, showing that, similar to HIPSTR, the expression of thousands of lncRNAs is more highly heterogeneous than the expression of mRNAs in the individual, otherwise indistinguishable cells of totipotent human embryos, primordial germ cells, and stable cell lines.
Collapse
|
19
|
Shi D, Xiao X, Tian Y, Qin L, Xie F, Sun R, Wang J, Li W, Liu T, Xiao Y, Yu W, Guo W, Xiong Y, Qiu H, Kang T, Huang W, Zhao C, Deng W. Activating enhancer-binding protein-2α induces cyclooxygenase-2 expression and promotes nasopharyngeal carcinoma growth. Oncotarget 2016; 6:5005-21. [PMID: 25669978 PMCID: PMC4467130 DOI: 10.18632/oncotarget.3215] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 12/26/2014] [Indexed: 12/29/2022] Open
Abstract
Activating enhancer-binding protein-2α (AP-2α) regulates the expression of many cancer-related genes. Here, we demonstrated a novel mechanism by which AP-2α up-regulated cyclooxygenase-2 (COX-2) expression to promote the growth of nasopharyngeal carcinomas (NPCs). High expression of AP-2α in NPC cell lines and tumor tissues from NPC patients was detected and significantly correlated with COX-2 expression. Overexpression of AP-2α and COX-2 in tumor tissues was associated with advanced tumor stage, clinical progression, and short survival of patients with NPCs. Knockdown of AP-2α by siRNA markedly inhibited COX-2 expression and PGE2 production in NPC cells. Exogenous expression of AP-2α up-regulated the COX-2 and PGE2. Knockdown of AP-2α also significantly suppressed cell proliferation in NPC cells in vitro and tumor growth in a NPC xenograft mouse model. Moreover, we found that p300 played an important role in the AP-2α/COX-2 pathway. AP-2α could co-localize and interact with p300 in NPC cells. Overexpression of the p300, but not its histone acetyltransferase (HAT) domain deletion mutant, promoted the acetylation of AP-2α and its binding on the COX-2 promoter, thereby up-regulated COX-2 expression. Our results indicate that AP-2α activates COX-2 expression to promote NPC growth and suggest that the AP-2α/COX-2 signaling is a potential therapeutic target for NPC treatment.
Collapse
Affiliation(s)
- Dingbo Shi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yun Tian
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Lijun Qin
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fangyun Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Rui Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jingshu Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenbin Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Tianze Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yao Xiao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuqing Xiong
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Huijuan Qiu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Chong Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| |
Collapse
|
20
|
Huang W, Chen C, Liang Z, Qiu J, Li X, Hu X, Xiang S, Ding X, Zhang J. AP-2α inhibits hepatocellular carcinoma cell growth and migration. Int J Oncol 2016; 48:1125-34. [PMID: 26780928 DOI: 10.3892/ijo.2016.3318] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/09/2015] [Indexed: 11/06/2022] Open
Abstract
Transcription factor AP-2α is involved in many types of human cancers, but its role in hepatocellular carcinogenesis is largely unknown. In this study, we found that expression of AP-2α was low in 40% of human hepatocellular cancers compared with adjacent normal tissues by immunohistochemical analysis. Moreover, AP-2α expression was low or absent in hepatocellular cancer cell lines (HepG2, Hep3B, SMMC-7721 and MHHC 97-H). Human liver cancer cell lines SMMC-7721 and Hep3B stably overexpressing AP-2α were established by lentiviral infection and puromycin screening, and the ectopic expression of AP-2α was able to inhibit hepatocellular cancer cell growth and proliferation by cell viability, MTT assay and liquid colony formation in vitro and in vivo. Furthermore, AP-2α overexpression decreased liver cancer cell migration and invasion as assessed by wound healing and Transwell assays, increasing the sensitivity of liver cancer cells to cisplatin analyzed by MTT assays. Also AP-2α overexpression suppressed the sphere formation and renewed the ability of cancer stem cells. Finally, we found that AP-2α is epigenetically modified and modulates the levels of phosphorylated extracellular signal-regulated protein kinase (ERK), β-catenin, p53, EMT, and CD133 expression in liver cancer cell lines. These results suggested that AP-2α expression is low in human hepatocellular cancers by regulating multiple signaling to affect hepatocellular cancer cell growth and migration. Therefore, AP-2α might represent a novel potential target in human hepatocellular cancer therapy.
Collapse
Affiliation(s)
- Wenhuan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Cheng Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhongheng Liang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Junlu Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xinxin Li
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
21
|
Sharma P, Sharma R. miRNA-mRNA crosstalk in esophageal cancer: From diagnosis to therapy. Crit Rev Oncol Hematol 2015; 96:449-62. [PMID: 26257289 DOI: 10.1016/j.critrevonc.2015.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 04/11/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022] Open
Abstract
The asymptomatic nature of esophageal cancer (EC) at early stages results in late clinical presentation leading to poor prognosis and limited success of therapeutic modalities. Efforts to identify diagnostic/prognostic markers have proven to be unsuccessful for translation into clinics. Hence, there is a pressing need for establishment of novel non-invasive biomarker for early diagnosis/better prognosis of EC. Recently, alteration in microRNA (miRNA) expression has emerged as an important hallmark of cancer. This review summarizes the differential expression of miRNAs in EC and addresses how their aberrant expression influences crucial biological processes such as apoptosis, cell proliferation, invasion and metastasis. Additionally, this review highlights the current status of circulating miRNA based diagnostic/prognostic markers. An effort has been made to find a connection between different miRNAs involved in EC and a detailed analysis has been done to screen out micoRNAs involved in prognosis and multidrug resistance. Further, investigation of these miRNAs would not only provide a gene therapy based strategy to prevent/treat cancer but also to reverse multidrug resistance leading to decreased requirement of harmful chemotherapeutic drugs.
Collapse
Affiliation(s)
- Priyanka Sharma
- Research Scholar, University School of Biotechnology, Guru Gobind Singh Indraprastha University, New Delhi 110078, India.
| | - Rinu Sharma
- Assistant Professor, University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector 16C Dwarka, New Delhi 110078, India.
| |
Collapse
|
22
|
Krušlin B, Ulamec M, Tomas D. Prostate cancer stroma: an important factor in cancer growth and progression. Bosn J Basic Med Sci 2015; 15:1-8. [PMID: 26042506 PMCID: PMC4469930 DOI: 10.17305/bjbms.2015.449] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 12/30/2022] Open
Abstract
Reactive stromal changes that occur in different human cancers might play a role in local tumor spreading and progression. Studies done on various human cancers have shown activated stromal cell phenotypes, modified extracellular matrix (ECM) composition, and increased microvessel density. Furthermore, they exhibit biological markers consistent with stroma at the site of wound repair. In prostate cancer, stroma is composed of fibroblasts, myofibroblasts, endothelial cells and immune cells. Predominant cells in the tumorous stroma are, however, fibroblasts/myofibroblasts. They are responsible for the synthesis, deposition and remodeling of the ECM. Epithelial tumorous cells, in interaction with stromal cells and with the help of various molecules of ECM, create a microenvironment suitable for cancer cell proliferation, movement, and differentiation. In this review, we discussed the role of different stromal components in prostate cancer as well as their potential prognostic and therapeutic significance.
Collapse
Affiliation(s)
- Božo Krušlin
- Department of pathology, Sestre milosrdnice University Hospital, Zagreb.
| | | | | |
Collapse
|
23
|
Sun J, Du N, Li J, Zhou J, Tao G, Sun S, He J. Transcription Factor AP2ε: A Potential Predictor of Chemoresistance in Patients With Gastric Cancer. Technol Cancer Res Treat 2015; 15:285-95. [PMID: 25810491 DOI: 10.1177/1533034615577028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/09/2015] [Indexed: 01/14/2023] Open
Abstract
Chemotherapy is a mainstay of therapy for advanced gastric cancer (GC); however, owing to drug resistances, the effectiveness of chemotherapy is not satisfactory for some patients with GC. Therefore, identification of a marker that predicts treatment response is beneficial to patients. Hypermethylation of transcription factor activating enhancer-binding protein 2∊ (TFAP2E) has been implicated in chemotherapy resistance to fluorouracil-based chemotherapy in patients with colorectal cancer, but its role in GC is still unknown. In this study, we investigated TFAP2E as a predictor of treatment response in GC. We used methylation-sensitive high-resolution melting analysis to study the methylation of TFAP2E in 141 GC tissue specimens and 45 adjacent nontumor tissue specimens. In vitro experiments, we analyzed the expression and methylation of TFAP2E and to examine the sensitivity of GC cell lines to 5-fluorouracil (5-FU). The TFAP2E methylation occurred at a significantly higher incidence rate in tumor tissues compared to adjacent nontumor tissues (chi-square [χ2] = 38.919, P < .001). Hypermethylation of TFAP2E occurred more frequently in tumors with lower differentiation grades (P < .001) and was significantly associated with nonresponse to fluorouracil-based chemotherapy (P = .010). Hypermethylation was also associated with decreased expression of TFAP2E (P < .01) and nonresponse to 5-FU exposure in vitro (P < .001). Hypermethylation of TFAP2E was associated with lack of response to fluorouracil-based chemotherapy, indicating that it might be a potential predictor of treatment response in patients with GC.
Collapse
Affiliation(s)
- Jingyue Sun
- Department of Oncology, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Nan Du
- Department of Oncology, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jin Li
- Department of Oncology, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Guoquan Tao
- Department of Gastrointestinal surgery, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Suan Sun
- Department of Pathology, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Jingdong He
- Department of Oncology, The Huai'an First People's Hospital, Affiliated to Nanjing Medical University, Huai'an, Jiangsu Province, China
| |
Collapse
|
24
|
Zhou L, Jang KY, Moon YJ, Wagle S, Kim KM, Lee KB, Park BH, Kim JR. Leptin ameliorates ischemic necrosis of the femoral head in rats with obesity induced by a high-fat diet. Sci Rep 2015; 5:9397. [PMID: 25797953 PMCID: PMC4369691 DOI: 10.1038/srep09397] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/03/2015] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for ischemic necrosis of the femoral head (INFH). The purpose of this study was to determine if leptin treatment of INFH stimulates new bone formation to preserve femoral head shape in rats with diet-induced obesity. Rats were fed a high-fat diet (HFD) or normal chow diet (NCD) for 16 weeks to induce progressive development of obesity. Avascular necrosis of the femoral head (AVN) was surgically induced. Adenovirus-mediated introduction of the leptin gene was by intravenous injection 2 days before surgery-induced AVN. At 6 weeks post-surgery, radiologic and histomorphometric assessments were performed. Leptin signaling in tissues was examined by Western blot. Osteogenic markers were analyzed by real-time RT-PCR. Radiographs showed better preservation of femoral head architecture in the HFD-AVN-Leptin group than the HFD-AVN and HFD-AVN-LacZ groups. Histology and immunohistochemistry revealed the HFD-AVN-Leptin group had significantly increased osteoblastic proliferation and vascularity in infarcted femoral heads compared with the HFD-AVN and HFD-AVN-LacZ groups. Intravenous injection of leptin enhanced serum VEGF levels and activated HIF-1α pathways. Runx 2 and its target genes were significantly upregulated in the HFD-AVN-Leptin group. These results indicate that leptin resistance is important in INFH pathogenesis. Leptin therapy could be a new strategy for INFH.
Collapse
Affiliation(s)
- Lu Zhou
- 1] Department of Orthopaedic Surgery, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea [2] Department of Sports Medicine, Taishan Medical University, Shandong, China
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Young Jae Moon
- Department of Biochemistry, Chonbuk National University Medical School, Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Sajeev Wagle
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Kwang Bok Lee
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Byung-Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Research Institute for Endocrine Sciences, Jeonju, Republic of Korea
| | - Jung Ryul Kim
- Department of Orthopaedic Surgery, Chonbuk National University Medical School, Research Institute for Endocrine Sciences and Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| |
Collapse
|
25
|
Visser M, Palstra RJ, Kayser M. Allele-specific transcriptional regulation of IRF4 in melanocytes is mediated by chromatin looping of the intronic rs12203592 enhancer to the IRF4 promoter. Hum Mol Genet 2015; 24:2649-61. [PMID: 25631878 DOI: 10.1093/hmg/ddv029] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/24/2015] [Indexed: 01/07/2023] Open
Abstract
The majority of significant single-nucleotide polymorphisms (SNPs) identified with genome-wide association studies are located in non-coding regions of the genome; it is therefore possible that they are involved in transcriptional regulation of a nearby gene rather than affecting an encoded protein's function. Previously, it was demonstrated that the SNP rs12203592, located in intron 4 of the IRF4 gene, is strongly associated with human skin pigmentation and modulates an enhancer element that controls expression of IRF4. In our study, we investigated the allele-specific effect of rs12203592 on IRF4 expression in epidermal skin samples and in melanocytic cells from donors of different skin color. We focused on the characteristics and activity of the enhancer, and on long-range chromatin interactions in melanocytic cells homozygous and heterozygous for rs12203592. We found that, irrespective of the trans-activating environment, IRF4 transcription is strongly correlated with the allelic status of rs12203592, the activity of the rs12203592 enhancer and that the chromatin features depend on the rs12203592 genotype. Furthermore, we demonstrate that the rs12203592 enhancer physically interacts with the IRF4 promoter through an allele-dependent chromatin loop, and suggest that subsequent allele-specific activation of IRF4 transcription is stabilized by another allele-specific loop from the rs12203592 enhancer to an additional regulatory element in IRF4. We conclude that the non-coding SNP rs12203592 is located in a regulatory region and affects a wide range of enhancer characteristics, resulting into modulation of the enhancer's activity, its interaction with the IRF4 promoter and subsequent allele-specific transcription of IRF4. Our findings provide another example of a non-coding SNP affecting skin color by modulating enhancer-mediated transcriptional regulation.
Collapse
Affiliation(s)
- Mijke Visser
- Department of Forensic Molecular Biology, Erasmus MC University Medical Centre Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Robert-Jan Palstra
- Department of Forensic Molecular Biology, Erasmus MC University Medical Centre Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Manfred Kayser
- Department of Forensic Molecular Biology, Erasmus MC University Medical Centre Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
26
|
Zhang ZM, Wang Y, Huang R, Liu YP, Li X, Hu FL, Zhu L, Wang F, Cui BB, Dong XS, Zhao YS. TFAP2E hypermethylation was associated with survival advantage in patients with colorectal cancer. J Cancer Res Clin Oncol 2014; 140:2119-27. [PMID: 24996990 DOI: 10.1007/s00432-014-1766-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/26/2014] [Indexed: 12/20/2022]
Abstract
PURPOSE Hypermethylation of TFAP2E (AP-2E) is associated with the chemotherapy-resistant in patients with colorectal cancer (CRC), but its implications on prognosis directly remain unknown. This study was aimed to investigate the role of AP-2E methylation status and other clinicopathologic parameters as predictors of prognosis. METHODS We detected the methylation status of AP-2E in tumor and adjacent non-tumor tissues from 311 sporadic CRC patients by methylation-sensitive high-resolution melting analysis. Log-rank tests and multivariate Cox analyses were performed to evaluate the role of AP-2E methylation status and other clinicopathologic parameters as predictors of prognosis. RESULTS Hypermethylation of AP-2E was detected in 61 % (190/311) tumor tissues. It occurred more frequently in tumors in earlier stages (I/II; P = 0.02), lower levels of tumor invasion (T1-T3; P = 0.04), fewer lymph nodes involved (N0; P < 0.01), and higher histologic grades (G1/G2; P < 0.01). The overall 5-year survival rates in hypermethylation and hypomethylation group were 76.91 and 47.17 % (P < 0.0001), respectively. AP-2E hypermethylation was significantly associated with a favorable clinical outcome with a hazard ratio of 0.486 (95 % CI 0.342-0.692, P < 0.0001) after controlling for age, gender, tumor location, histologic type, TNM staging, and histologic grade. CONCLUSIONS AP-2E was frequently hypermethylated in tumors from patients with CRC. Aberrant hypermethylation of AP-2E occurred more frequently in tumors with earlier stages, lower levels of tumor invasion, fewer lymph nodes involved, and higher histologic grades. AP-2E hypermethylation might be an independent predictor of survival advantage in patients with CRC.
Collapse
Affiliation(s)
- Zuo-Ming Zhang
- Department of Epidemiology, School of Public Health, Harbin Medical University, 157 Baojian Road, Harbin, 150086, Heilongjiang Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Meng X, Meng C, Yang B, Zhao L, Sun X, Su Y, Liu H, Fan F, Liu X, Jia L. AP-2α downregulation by cigarette smoke condensate is counteracted by p53 in human lung cancer cells. Int J Mol Med 2014; 34:1094-100. [PMID: 25050743 DOI: 10.3892/ijmm.2014.1857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 07/10/2014] [Indexed: 11/06/2022] Open
Abstract
Cumulative findings have demonstrated that the dysregulation of tumor suppressor genes may be implicated in cigarette smoke-induced carcinogenesis. Activating enhancer-binding protein 2 (AP-2) is a eukaryotic transcriptional factor that plays a significant role in embryonic development and tumorigenesis. The vertebrate AP-2 family consists of AP-2α, AP-2β, AP-2γ, AP-2δ and AP-2ε. Previous studies have suggested that cigarette smoking disrupts AP-2 regulation. In the present study, we investigated the effects of cigarette smoke condensate (CSC) on AP-2α expression in human lung cancer cell lines (NCI-H1299, NCI-H446 and A549), as well as the potential mechanisms involved. Using RT-qPCR, we found that CSC decreased AP-2α expression by suppressing its transcription in human lung cancer cell lines, particularly in p53-deficient NCI-H1299 cells. Western blotting and luciferase assays were implemented and we found that the restoration of p53 expression rescued the NCI-H1299 cells from CSC-induced AP-2α loss, while the silencing of p53 resulted in increased AP-2α loss induced by CSC, suggesting an antagonizing role of p53 in the regulation of AP-2α by CSC. Our results indicate that AP-2α downregulation may be involved in smoke-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Xiangjun Meng
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Cuida Meng
- Department of Otolaryngology Head and Neck Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bing Yang
- Department of Cell Biology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Li Zhao
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuefei Sun
- Department of Emergency, China-Japan Union Hospital, Changchun, Jilin 130021, P.R. China
| | - Yun Su
- Department of Orthopedics, Affiliated Zhongshan Hospital, Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Hongyang Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Feiyue Fan
- Department of Radiation Hazard Evaluation, Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192, P.R. China
| | - Xiaodong Liu
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lili Jia
- Key Laboratory of Radiobiology (Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
28
|
Motalleb G, Gholipour N, Samaei NM. Association of the human astrocyte elevated gene-1 promoter variants with susceptibility to hepatocellular carcinoma. Med Oncol 2014; 31:916. [PMID: 24659263 DOI: 10.1007/s12032-014-0916-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/07/2014] [Indexed: 12/20/2022]
Abstract
Central role of astrocyte elevated gene-1 (AEG-1) in regulating diverse aspects of hepatocellular carcinoma (HCC) pathogenesis and association of its overexpression with HCC progression has been demonstrated. The positive regulatory regions of AEG-1 promoter contain several putative transcription factor binding sites critical for basal promoter activity. In this study, the aim was to explore the association of AEG-1 promoter variant with HCC. In this study, the human AEG-1 promoter including the region -538 to -42 was explored in 53 HCC patients and 108 healthy controls. The polymerase chain reaction-sequencing method was used for investigating AEG-1 promoter polymorphisms. A novel mutation in AEG-1 promoter in human HCC patients at a potential AP-2 binding site was explored. An A>C mutation was observed in -483 of AEG-1 promoter in 4 out of 53 HCC patients but not in 108 control individuals. Sequencing data showed genetic variations in 11 HCC patients and 3 healthy controls. Among them, one novel SNP was found in activator protein-1 (AP2), a transcription factor binding site (-483 A to C) that may be associated with the susceptibility to HCC (P = 0.012) but no associations were found for other observed variations. This mutation could be tumor-specific. AEG-1 promoter variant -483 A>C may be associated with the susceptibility to HCC in Iranian population. To our knowledge, this is the first study that has reported this association with the susceptibility to HCC. Therefore, further studies need to be conducted in larger sample sizes and other populations to validate these findings.
Collapse
Affiliation(s)
- Gholamreza Motalleb
- Department of Biology, Faculty of Science, University of Zabol, P.O. Box 98615-53, Zabol, Iran,
| | | | | |
Collapse
|
29
|
Shi D, Xie F, Zhang Y, Tian Y, Chen W, Fu L, Wang J, Guo W, Kang T, Huang W, Deng W. TFAP2A Regulates Nasopharyngeal Carcinoma Growth and Survival by Targeting HIF-1α Signaling Pathway. Cancer Prev Res (Phila) 2013; 7:266-77. [DOI: 10.1158/1940-6207.capr-13-0271] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Ding X, Yang Z, Zhou F, Wang F, Li X, Chen C, Li X, Hu X, Xiang S, Zhang J. Transcription factor AP-2α regulates acute myeloid leukemia cell proliferation by influencing Hoxa gene expression. Int J Biochem Cell Biol 2013; 45:1647-56. [DOI: 10.1016/j.biocel.2013.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/26/2013] [Accepted: 04/29/2013] [Indexed: 01/28/2023]
|
31
|
MacDonald ST, Bamforth SD, Bragança J, Chen CM, Broadbent C, Schneider JE, Schwartz RJ, Bhattacharya S. A cell-autonomous role of Cited2 in controlling myocardial and coronary vascular development. Eur Heart J 2013; 34:2557-2565. [PMID: 22504313 PMCID: PMC3748368 DOI: 10.1093/eurheartj/ehs056] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 01/30/2012] [Accepted: 02/16/2012] [Indexed: 02/06/2023] Open
Abstract
AIMS Myocardial development is dependent on concomitant growth of cardiomyocytes and a supporting vascular network. The coupling of myocardial and coronary vascular development is partly mediated by vascular endothelial growth factor (VEGFA) signalling and additional unknown mechanisms. We examined the cardiomyocyte specific role of the transcriptional co-activator Cited2 on myocardial microstructure and vessel growth, in relation to Vegfa expression. METHODS AND RESULTS A cardiomyocyte-specific knockout of mouse Cited2 (Cited2(Nkx)) was analysed using magnetic resonance imaging and histology. Ventricular septal defects and significant compact layer thinning (P < 0.02 at right ventricular apex, P < 0.009 at the left ventricular apex in Cited2(Nkx) vs. controls, n = 11 vs. n = 7, respectively) were found. This was associated with a significant decrease in the number of capillaries to larger vessels (ratio 1.56 ± 0.56 vs. 3.25 ± 1.63, P = 2.7 × 10(-6) Cited2(Nkx) vs. controls, n = 11 vs. n = 7, respectively) concomitant with a 1.5-fold reduction in Vegfa expression (P < 0.02, Cited2(Nkx) vs. controls, n = 12 vs. n = 12, respectively). CITED2 was subsequently found at the Vegfa promoter in mouse embryonic hearts using chromatin immunoprecipitation, and moreover found to stimulate human VEGFA promoter activity in cooperation with TFAP2 transcription factors in transient transfection assays. There was no change in the myocardial expression of the left-right patterning gene Pitx2c, a previously known target of CITED2. CONCLUSIONS This study delineates a novel cell-autonomous role of Cited2 in regulating VEGFA transcription and the development of myocardium and coronary vasculature in the mouse. We suggest that coupling of myocardial and coronary growth in the developing heart may occur in part through a Cited2→Vegfa pathway.
Collapse
Affiliation(s)
- Simon T. MacDonald
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Simon D. Bamforth
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - José Bragança
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Chiann-Mun Chen
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Carol Broadbent
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Jürgen E. Schneider
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| | - Robert J. Schwartz
- Institute of Biosciences and Technology, Texas A&M Health Science Centre, Houston, TX 77030-3498, USA
| | - Shoumo Bhattacharya
- Department of Cardiovascular Medicine, University of Oxford and Wellcome Trust Centre for Human Genetics, Roosevelt Drive, OxfordOX3 7BN, UK
| |
Collapse
|
32
|
Lin HH, Chen YH, Chiang MT, Huang PL, Chau LY. Activator protein-2α mediates carbon monoxide-induced stromal cell-derived factor-1α expression and vascularization in ischemic heart. Arterioscler Thromb Vasc Biol 2013; 33:785-94. [PMID: 23393395 DOI: 10.1161/atvbaha.112.301143] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Increased cardiac stromal cell-derived factor-1α (SDF-1α) expression promotes neovascularization and myocardial repair after ischemic injury through recruiting stem cells and reducing cardiomyocyte death. Previous studies have shown that heme oxygenase-1 and its reaction byproduct, carbon monoxide (CO), induce SDF-1α expression in ischemic heart. However, the mechanism underlying heme oxygenase-1/CO-induced cardiac SDF-1α expression remains elusive. This study aims to investigate the signaling pathway and the transcriptional factor that mediate CO-induced SDF-1α gene expression and cardioprotection. APPROACH AND RESULTS CO gas and a CO-releasing compound, tricarbonyldichlororuthenium (II) dimer, dose-dependently induced SDF-1α expression in primary neonatal cardiomyocytes and H9C2 cardiomyoblasts. Promoter luciferase-reporter assay, electrophoretic mobility shift assay, and chromatin immunoprecipitation demonstrated that the activator protein 2α (AP-2α) mediated tricarbonyldichlororuthenium (II) dimer-induced SDF-1α gene transcription. Tricarbonyldichlororuthenium (II) dimer induced AP-2α expression via protein kinase B (AKT)-dependent signaling. AKT inhibition or AP-2α knockdown reduced tricarbonyldichlororuthenium (II) dimer-induced SDF-1α expression. Coronary ligation induced transient increases of cardiac AP-2α and SDF-1α expression, which were declined at 1 week postinfarction in mice. Periodic exposure of coronary-ligated mice to CO (250 ppm for 1 hour/day, 6 days) resumed the induction of AP-2α and SDF-1α gene expression in infarcted hearts. Immunohistochemistry and echocardiography performed at 4 weeks after coronary ligation revealed that CO treatment enhanced neovascularization in the myocardium of peri-infarct region and improved cardiac function. CO-mediated SDF-1α expression and cardioprotection was ablated by intramyocardial injection of lentivirus bearing specific short hairpin RNA targeting AP-2α. CONCLUSIONS Our data demonstrate that AKT-dependent upregulation of AP-2α is essential for CO-induced SDF-1α expression and myocardial repair after ischemic injury.
Collapse
Affiliation(s)
- Heng-Huei Lin
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan, ROC
| | | | | | | | | |
Collapse
|
33
|
Eisermann K, Broderick CJ, Bazarov A, Moazam MM, Fraizer GC. Androgen up-regulates vascular endothelial growth factor expression in prostate cancer cells via an Sp1 binding site. Mol Cancer 2013; 12:7. [PMID: 23369005 PMCID: PMC3616929 DOI: 10.1186/1476-4598-12-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 01/21/2013] [Indexed: 11/26/2022] Open
Abstract
Background Vascular Endothelial Growth Factor (VEGF) is regulated by a number of different factors, but the mechanism(s) behind androgen-mediated regulation of VEGF in prostate cancer are poorly understood. Results Three novel androgen receptor (AR) binding sites were discovered in the VEGF promoter and in vivo binding of AR to these sites was demonstrated by chromatin immunoprecipitation. Mutation of these sites attenuated activation of the VEGF promoter by the androgen analog, R1881 in prostate cancer cells. The transcription factors AR and Sp1 were shown to form a nuclear complex and both bound the VEGF core promoter in chromatin of hormone treated CWR22Rv1 prostate cancer cells. The importance of the Sp1 binding site in hormone mediated activation of VEGF expression was demonstrated by site directed mutagenesis. Mutation of a critical Sp1 binding site (Sp1.4) in the VEGF core promoter region prevented activation by androgen. Similarly, suppression of Sp1 binding by Mithramycin A treatment significantly reduced VEGF expression. Conclusions Our mechanistic study of androgen mediated induction of VEGF expression in prostate cancer cells revealed for the first time that this induction is mediated through the core promoter region and is dependent upon a critical Sp1 binding site. The importance of Sp1 binding suggests that therapy targeting the AR-Sp1 complex may dampen VEGF induced angiogenesis and, thereby, block prostate cancer progression, helping to maintain the indolent form of prostate cancer.
Collapse
Affiliation(s)
- Kurtis Eisermann
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | | | | | | | | |
Collapse
|
34
|
Synergistic silencing by promoter methylation and reduced AP-2α transactivation of the proapoptotic HRK gene confers apoptosis resistance and enhanced tumor growth. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:84-95. [PMID: 23159945 DOI: 10.1016/j.ajpath.2012.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 09/06/2012] [Accepted: 09/18/2012] [Indexed: 02/05/2023]
Abstract
The Harakiri (HRK) gene encodes an important proapoptotic mitochondrial protein of the Bcl-2 family. HRK is expressed in normal tissues but is decreased in many cancers such as melanoma, the mechanisms of which have not been fully elucidated. Here, we demonstrate that HRK is silenced by hypermethylation of a major proximal CpG island in the HRK promoter. Furthermore, we show that HRK is a novel target gene regulated by the transcription factor AP-2α, which interacts with an AP-2α binding site in the HRK promoter. Hypermethylation of the major proximal CpG island (which contains the AP-2α binding site within the most densely methylated -218- to -194-bp region) inhibited AP-2α binding and transcriptional activity. Artificial overexpression of AP-2α in melanoma cells up-regulated HRK transcription, which was further restored by treatment with DNA methyltransferase inhibitor 5-azacytidine. Artificial overexpression of HRK by recombinant adenovirus induced caspase-dependent apoptosis, inhibited melanoma cell growth in vitro, and markedly reduced in vivo melanoma growth in a nude mouse xenograft model. RNA interference by siHRK or siAP-2α reversed the above effects. We conclude that the synergistic effects of HRK promoter hypermethylation and loss of AP-2α transactivation lead to HRK gene silencing and confer resistance to apoptosis and enhanced tumor growth. These novel molecular lesions may provide the basis for new therapeutic approaches to treating AP-2α- and HRK-deficient cancers.
Collapse
|
35
|
AP-2α–dependent regulation of Bcl-2/Bax expression affects apoptosis in the trophoblast. J Mol Histol 2012; 43:681-9. [DOI: 10.1007/s10735-012-9439-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
|
36
|
|
37
|
Chen C, Guo L, Shi M, Hu M, Hu M, Yu M, Wang T, Song L, Shen B, Qian L, Guo N. Modulation of IFN-γ receptor 1 expression by AP-2α influences IFN-γ sensitivity of cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:661-71. [PMID: 22182699 DOI: 10.1016/j.ajpath.2011.10.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/18/2011] [Accepted: 10/31/2011] [Indexed: 12/25/2022]
Abstract
Interferon (IFN)-γ plays crucial roles in regulating both innate and adaptive immunity. The existence of IFN-γ receptor 1 (IFNGR1) molecules on the cell surface is a prerequisite to the initiation of IFN-γ signaling; low expression of IFNGR1 leads to a functional blockade of IFN-γ signaling. However, the molecular mechanisms by which IFNGR1 expression is controlled are unclear. In the present study, we demonstrated that IFNGR1 expression was reduced or lost in breast cancer. Heterogeneous IFNGR1 immunoreactivity appeared to be associated with the morphological heterogeneity of breast cancer, and loss of IFNGR1 expression was predominantly observed in poorly differentiated areas. We identified the functional activating protein (AP)-2 and specificity protein (SP)-1 sites within the IFNGR1 promoter. Ectopic expression of AP-2α drastically repressed the expression of IFNGR1 and hindered IFN-γ signaling, whereas AP-2α gene silencing elevated IFNGR1 levels. Overexpression of SP-1 effectively antagonized the repressive effects of AP-2α. Simultaneous recruitment of both transcription factors to the AP-2 and SP-1 motifs, respectively, in the IFNGR1 promoter was demonstrated, implying that AP-2α and SP-1 may synergistically modulate IFNGR1 transcription. Moreover, AP-2α overexpression in AP-2-deficient SW480 cells remarkably inhibited Stat1 phosphorylation and the anti-proliferative effects of IFN-γ, whereas knockdown of the AP-2α expression dramatically enhanced the sensitivities of HeLa cells highly expressing AP-2 to IFN-γ, indicating that dysregulation of AP-2α expression is associated with impaired IFN-γ actions in cancer cells.
Collapse
Affiliation(s)
- Changguo Chen
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wu Y, Xiao Y, Ding X, Zhuo Y, Ren P, Zhou C, Zhou J. A miR-200b/200c/429-binding site polymorphism in the 3' untranslated region of the AP-2α gene is associated with cisplatin resistance. PLoS One 2011; 6:e29043. [PMID: 22194984 PMCID: PMC3237583 DOI: 10.1371/journal.pone.0029043] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 11/18/2011] [Indexed: 01/13/2023] Open
Abstract
The transcription factor AP-2α functions as a tumor suppressor by regulating various genes that are involved in cell proliferation and apoptosis. Chemotherapeutic drugs including cisplatin induce post-transcriptionally endogenous AP-2α, which contributes to chemosensitivity by enhancing therapy-induced apoptosis. microRNAs (miRNAs) miR-200b, miR-200c and miR-429 (miR-200b/200c/429) are up-regulated in endometrial and esophageal cancers, and their overexpression correlates with resistance to cisplatin treatment. Using computational programs, we predicted that the 3′ untranslated region (UTR) of AP-2α gene contains a potential miRNA response element (MRE) for the miR-200b/200c/429 family, and the single nucleotide polymorphism (SNP) site rs1045385 (A or C allele) resided within the predicted MRE. Luciferase assays and Western blot analysis demonstrated that the miR-200b/200c/429 family recognized the MRE in the 3′ UTR of AP-2α gene and negatively regulated the expression of endogenous AP-2α proteins. SNP rs1045385 A>C variation enhanced AP-2α expression by disrupting the binding of the miR-200b/200c/429 family to the 3′ UTR of AP-2α. The effects of the two polymorphic variants on cisplatin sensitivity were determined by clonogenic assay. The overexpression of AP-2α with mutant 3′ UTR (C allele) in the endometrial cancer cell line HEC-1A, which has high levels of endogenous miR-200b/200c/429 and low levels of AP-2α protein, significantly increased cisplatin sensitivity, but overexpression of A allele of AP-2α has no significant effects, compared with mock transfection. We concluded that miR-200b/200c/429 induced cisplatin resistance by repressing AP-2α expression in endometrial cancer cells. The SNP (rs1045385) A>C variation decreased the binding of miR-200b/200c/429 to the 3′ UTR of AP-2α, which upregulated AP-2α protein expression and increased cisplatin sensitivity. Our results suggest that SNP (rs1045385) may be a potential prognostic marker for cisplatin treatment.
Collapse
Affiliation(s)
- Yuan Wu
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
| | - Yuzhong Xiao
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
| | - Xiaofeng Ding
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
| | - Yiming Zhuo
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
| | - Peng Ren
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
| | - Chang Zhou
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
- * E-mail: (JZ); (CZ)
| | - Jianlin Zhou
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Science, Hunan Normal University, Changsha, China
- * E-mail: (JZ); (CZ)
| |
Collapse
|
39
|
Wang W, Lv L, Pan K, Zhang Y, Zhao JJ, Chen JG, Chen YB, Li YQ, Wang QJ, He J, Chen SP, Zhou ZW, Xia JC. Reduced expression of transcription factor AP-2α is associated with gastric adenocarcinoma prognosis. PLoS One 2011; 6:e24897. [PMID: 21966377 PMCID: PMC3180284 DOI: 10.1371/journal.pone.0024897] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/19/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND This study aims to investigate the expression and prognostic significance of activator protein 2α (AP-2α) in gastric adenocarcinoma. METHODOLOGY/PRINCIPAL FINDINGS AP-2α expression was analyzed using real-time quantitative PCR (RT-qPCR), western blotting, and immunohistochemical staining methods on tissue samples from a consecutive series of 481 gastric adenocarcinoma patients who underwent resections between 2003 and 2006. The relationship between AP-2α expression, clinicopathological factors, and patient survival was investigated. RT- qPCR results showed that the expression of AP-2α mRNA was reduced in tumor tissue samples, compared with expression in matched adjacent non-tumor tissue samples (P = 0.009); this finding was confirmed by western blotting analysis (P = 0.012). Immunohistochemical staining data indicated that AP-2α expression was significantly decreased in 196 of 481 (40.7%) gastric adenocarcinoma cases; reduced AP-2α expression was also observed in patients with poorly differentiated tumors (P = 0.001) and total gastric carcinomas (P = 0.002), as well as in patients who underwent palliative tumor resection (P = 0.004). Additionally, reduced expression of AP-2α was more commonly observed in tumors that were staged as T4a/b (P = 0.018), N3 (P = 0.006), and M1 (P = 0.008). Kaplan-Meier survival curves revealed that reduced expression of AP-2α was associated with poor prognosis in gastric adenocarcinoma patients (P<0.001). Multivariate Cox analysis identified AP-2α expression as an independent prognostic factor for overall survival (HR = 1.512, 95% CI = 1.127-2.029, P = 0.006). CONCLUSIONS/SIGNIFICANCE Our data suggest that AP-2α plays an important role in tumor progression and that reduced AP-2α expression independently predicts an unfavorable prognosis in gastric adenocarcinoma patients.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Gastric and Pancreatic Surgery, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lin Lv
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ke Pan
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Pathology, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing-jing Zhao
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ju-gao Chen
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yi-bing Chen
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yong-qiang Li
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qi-jin Wang
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jia He
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shi-ping Chen
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-wei Zhou
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Gastric and Pancreatic Surgery, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- * E-mail: (ZZ-W); (XJ-C)
| | - Jian-chuan Xia
- State Key Laboratory of Oncology in South China and Department of Experimental Research, Cancer Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- * E-mail: (ZZ-W); (XJ-C)
| |
Collapse
|
40
|
Makhov PB, Golovine KV, Kutikov A, Canter DJ, Rybko VA, Roshchin DA, Matveev VB, Uzzo RG, Kolenko VM. Reversal of epigenetic silencing of AP-2alpha results in increased zinc uptake in DU-145 and LNCaP prostate cancer cells. Carcinogenesis 2011; 32:1773-81. [PMID: 21940908 DOI: 10.1093/carcin/bgr212] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Zinc accumulation is lost during prostate carcinogenesis. Recent studies reveal a strong association between prostate cancer progression and the downregulation of the zinc uptake transporters hZip1 and hZip3. The aim of this work was to assess the involvement of epigenetic processes in the disruption of zinc uptake homeostasis in prostate adenocarcinoma. In this report, we demonstrate an increase in hZip1 and hZip3 zinc transporters' expression and zinc uptake by the prostate cancer cells DU-145 and LNCaP in response to 5-aza-2'-deoxycytidine. This effect is due to demethylation of the promoter region of the activator protein (AP)-2alpha protein, which is crucial for hZip1 and hZip3 genes expression. Loss of AP-2alpha expression in DU-145 and LNCaP prostate cancer cells is due to hypermethylation of its promoter region. Similarly, we found higher AP-2alpha promoter methylation levels in clinical samples of early-stage prostate adenocarcinoma when compared with adjacent non-malignant prostate tissue. Taken together, our findings provide a better understanding of the epigenetic mechanisms that are involved in the loss of AP-2alpha protein in prostate cancer cells which lead to decreased cellular zinc uptake-a sine qua non of prostate cancer development.
Collapse
Affiliation(s)
- Peter B Makhov
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zou L, Sun Y, Wang M, Zhan Q. Aurora-A interacts with AP-2α and down regulates its transcription activity. PLoS One 2011; 6:e23110. [PMID: 21829699 PMCID: PMC3148253 DOI: 10.1371/journal.pone.0023110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 07/06/2011] [Indexed: 11/18/2022] Open
Abstract
Aurora-A is a serine/threonine protein kinase and plays an important role in the control of mitotic progression. Dysregulated expression of Aurora-A impairs centrosome separation and maturation, which lead to disrupted cell cycle progression and tumorigenesis. However, the molecular mechanism by which Aurora-A causes cell malignant transformation remains to be further defined. In this report, using transcription factors array and mRNA expression profiling array, we found that overexpression of Aurora-A suppressed transcription activity of AP-2α, a tumor suppressor that is often downregulated in variety of tumors, and inhibited expression of AP-2α-regulated downstream genes. These array-based observations were further confirmed by microwell colorimetric TF assay and luciferase reporter assay. Downregulated transcription activity of AP-2α by Aurora-A was found to be associated with reduced AP-2α protein stability, which appeared to be mediated by Aurora-A enhanced ubiquitin-dependent proteasomal degradation of AP-2α protein. Interestingly, Aurora-A-mediated AP-2α degradation was likely dependent Aurora-A kinase activity since inhibition of Aurora-A kinase activity was able to rescue Aurora-A-induced degradation of AP-2α. Moreover, we defined a physical interaction between Aurora-A and AP-2α, and such interaction might bridge the suppressive effect of Aurora-A on AP-2α protein stability. These findings provide new insights into molecular mechanism by which Aurora-A acts as an oncogenic molecule in tumor occurrence and malignant development.
Collapse
Affiliation(s)
- Lihui Zou
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yimin Sun
- CapitalBio Corporation, Beijing, China
| | - Mingrong Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
42
|
Abstract
OBJECTIVES The present study was conducted to evaluate the expression and function of AP-2α isoforms in pancreatic ductal adenocarcinoma. METHODS The expression of AP-2α was evaluated at the RNA level by reverse transcription-polymerase chain reaction and at the protein level by Western blotting and immunofluorescence. Its function as a transcription factor was evaluated in transient transfection experiments: DNA binding properties by electromobility shift assay and transactivation capabilities by luciferase assay. RESULTS Multiple alternative splicing events of AP-2α messenger occurred in all human pancreatic cancer cell lines, including a novel isoform, termed variant 6, which was not present in HeLa cells. At the protein level, except for 1 cell line, all pancreatic cancer cell lines expressed high nuclear levels of AP-2α. We also showed that AP-2α expressed by the pancreatic cancer cell lines could bind its cognate recognition site and activate transcription. However, variant 6, although not able to activate transcription, did not act in a dominant negative manner when cotransfected with the full-length protein. CONCLUSIONS Multiple isoforms of AP-2α are highly expressed in pancreatic cancer cell lines including a new isoform, AP-2α variant 6, which seems to be pancreatic cancer specific and is deprived of transcriptional activity.
Collapse
|
43
|
AP-2α expression and cell apoptosis of the lung tissue of rats with COPD and ECV304 cells stimulated by cigarette smoke extract. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/s11434-011-4437-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
44
|
Braeuer RR, Zigler M, Villares GJ, Dobroff AS, Bar-Eli M. Transcriptional control of melanoma metastasis: the importance of the tumor microenvironment. Semin Cancer Biol 2010; 21:83-8. [PMID: 21147226 DOI: 10.1016/j.semcancer.2010.12.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 12/02/2010] [Indexed: 11/18/2022]
Abstract
The molecular changes associated with the transition of melanoma cells from radial growth phase (RGP) to vertical growth phase (VGP) and the metastatic phenotype are not very well defined. However, some of the genes involved in this process and their transcriptional regulation are beginning to be elucidated. For example, the switch from RGP to VGP and the metastatic phenotype is associated with loss of the AP-2α transcription factor. AP-2α regulates the expression of c-KIT, MMP-2, VEGF, and the adhesion molecule MCAM/MUC18. Recently, we reported that AP-2α also regulates two G-protein coupled receptors (GPCRs) PAR-1 and PAFR. In turn, the thrombin receptor, PAR-1, regulates the expression of the gap junction protein Connexin-43 and the tumor suppressor gene Maspin. Activation of PAR-1 also leads to overexpression and secretion of proangiogenic factors such as IL-8, uPA, VEGF, PDGF, as well certain integrins. PAR-1 also cooperates with PAFR to regulate the expression of the MCAM/MUC18 via phosphorylation of CREB. The ligands for these GPCRs, thrombin and PAF, are secreted by stromal cells, emphasizing the importance of the tumor microenvironment in melanoma metastasis. The metastatic phenotype of melanoma is also associated with overexpression and function of CREB/ATF-1. Loss of AP-2α and overexpression of CREB/ATF-1 results in the overexpression of MCAM/MUC18 which by itself contributes to melanoma metastasis by regulating the inhibitor of DNA binding-1 (Id-1). CREB/ATF-1 also regulates the angiogenic factor CYR-61. Our recent data indicate that CREB/ATF-1 regulates the expression of AP-2α, thus, supporting the notion that CREB is an important "master switch" in melanoma progression.
Collapse
Affiliation(s)
- Russell R Braeuer
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
45
|
Gravina GL, Festuccia C, Marampon F, Popov VM, Pestell RG, Zani BM, Tombolini V. Biological rationale for the use of DNA methyltransferase inhibitors as new strategy for modulation of tumor response to chemotherapy and radiation. Mol Cancer 2010; 9:305. [PMID: 21108789 PMCID: PMC3001713 DOI: 10.1186/1476-4598-9-305] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 11/25/2010] [Indexed: 12/31/2022] Open
Abstract
Epigenetic modifications play a key role in the patho-physiology of many tumors and the current use of agents targeting epigenetic changes has become a topic of intense interest in cancer research. DNA methyltransferase (DNMT) inhibitors represent a promising class of epigenetic modulators. Research performed yielded promising anti-tumorigenic activity for these agents in vitro and in vivo against a variety of hematologic and solid tumors. These epigenetic modulators cause cell cycle and growth arrest, differentiation and apoptosis. Rationale for combining these agents with cytotoxic therapy or radiation is straightforward since the use of DNMT inhibitor offers greatly improved access for cytotoxic agents or radiation for targeting DNA-protein complex. The positive results obtained with these combined approaches in preclinical cancer models demonstrate the potential impact DNMT inhibitors may have in treatments of different cancer types. Therefore, as the emerging interest in use of DNMT inhibitors as a potential chemo- or radiation sensitizers is constantly increasing, further clinical investigations are inevitable in order to finalize and confirm the consistency of current observations.The present article will provide a brief review of the biological significance and rationale for the clinical potential of DNMT inhibitors in combination with other chemotherapeutics or ionizing radiation. The molecular basis and mechanisms of action for these combined treatments will be discussed herein.
Collapse
Affiliation(s)
- Giovanni L Gravina
- Department of Experimental Medicine, Division of Radiation Oncology, S, Salvatore Hospital, L'Aquila, University of L'Aquila, Medical School, L'Aquila 67100, Italy.
| | | | | | | | | | | | | |
Collapse
|
46
|
Orso F, Corà D, Ubezio B, Provero P, Caselle M, Taverna D. Identification of functional TFAP2A and SP1 binding sites in new TFAP2A-modulated genes. BMC Genomics 2010; 11:355. [PMID: 20525283 PMCID: PMC2890567 DOI: 10.1186/1471-2164-11-355] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 06/03/2010] [Indexed: 12/20/2022] Open
Abstract
Background Different approaches have been developed to dissect the interplay between transcription factors (TFs) and their cis-acting sequences on DNA in order to identify TF target genes. Here we used a combination of computational and experimental approaches to identify novel direct targets of TFAP2A, a key TF for a variety of physiological and pathological cellular processes. Gene expression profiles of HeLa cells either silenced for TFAP2A by RNA interference or not were previously compared and a set of differentially expressed genes was revealed. Results The regulatory regions of 494 TFAP2A-modulated genes were analyzed for the presence of TFAP2A binding sites, employing the canonical TFAP2A Positional Weight Matrix (PWM) reported in Jaspar http://jaspar.genereg.net/. 264 genes containing at least 2 high score TFAP2A binding sites were identified, showing a central role in "Cellular Movement" and "Cellular Development". In an attempt to identify TFs that could cooperate with TFAP2A, a statistically significant enrichment for SP1 binding sites was found for TFAP2A-activated but not repressed genes. The direct binding of TFAP2A or SP1 to a random subset of TFAP2A-modulated genes was demonstrated by Chromatin ImmunoPrecipitation (ChIP) assay and the TFAP2A-driven regulation of DCBLD2/ESDN/CLCP1 gene studied in details. Conclusions We proved that our computational approaches applied to microarray selected genes are valid tools to identify functional TF binding sites in gene regulatory regions as confirmed by experimental validations. In addition, we demonstrated a fine-tuned regulation of DCBLD2/ESDN transcription by TFAP2A.
Collapse
Affiliation(s)
- Francesca Orso
- Molecular Biotechnology Center (MBC), Department of Oncological Sciences, University of Torino, Torino, Italy
| | | | | | | | | | | |
Collapse
|
47
|
Leptin upregulates VEGF in breast cancer via canonic and non-canonical signalling pathways and NFkappaB/HIF-1alpha activation. Cell Signal 2010; 22:1350-62. [PMID: 20466060 DOI: 10.1016/j.cellsig.2010.05.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 05/05/2010] [Indexed: 01/26/2023]
Abstract
High levels of VEGF and leptin are strongly linked to worse prognosis of breast cancer. Leptin signalling upregulates VEGF in human and mouse mammary tumor cells (MT), but the specific molecular mechanisms are largely unknown. Pharmacologic and genetic approaches were used to dissect the mechanism of leptin regulation of VEGF protein and mRNA in MT (4T1, EMT6 and MMT). A series of VEGF-promoter Luc-reporters (full-length and transcription factor-binding deletions) were transfected into MT to analyze leptin regulation of VEGF transcription. Deletion analysis of VEGF promoter and RNA knockdown shows that HIF-1alpha and NFkappaB are essentials for leptin regulation of VEGF. Leptin activation of HIF-1alpha was mainly linked to canonic (MAPK, PI-3K) and non-canonic (PKC, JNK and p38 MAP) signalling pathways. Leptin non-canonic signalling pathways (JNK, p38 MAP and to less extent PKC) were linked to NFkappaB activation. SP1 was involved in leptin regulation of VEGF in 4T1 cells. AP1 was not involved and AP2 repressed leptin-induced increase of VEGF. Overall, these data suggest that leptin signalling regulates VEGF mainly through HIF-1alpha and NFkappaB. These results delineate a comprehensive mechanism for leptin regulation of VEGF in MT. Disruption of leptin signalling could be used as a novel way to treat breast cancer.
Collapse
|
48
|
Do TN, Ucisik-Akkaya E, Davis CF, Morrison BA, Dorak MT. An intronic polymorphism of IRF4 gene influences gene transcription in vitro and shows a risk association with childhood acute lymphoblastic leukemia in males. Biochim Biophys Acta Mol Basis Dis 2009; 1802:292-300. [PMID: 19897031 DOI: 10.1016/j.bbadis.2009.10.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 10/27/2009] [Accepted: 10/28/2009] [Indexed: 02/07/2023]
Abstract
The interferon regulatory factor (IRF) family of DNA-binding proteins regulates expression of interferon-inducible genes with roles in the immune response and carcinogenesis. IRF4 is involved in the differentiation of B and T cells and is overexpressed in B-cell malignancies as a result of c-REL (NF-kappaB) hyperactivation. IRF4 polymorphisms are associated with susceptibility to chronic lymphoid leukemia (CLL) and non-Hodgkin lymphoma (NHL). We examined 13 IRF4 SNPs in 114 cases of childhood acute lymphoblastic leukemia (ALL) and 388 newborn controls from Wales (U.K.) using TaqMan assays. IRF4 intron 4 SNP rs12203592 showed a male-specific risk association (OR=4.4, 95% CI=1.5 to 12.6, P=0.007). Functional consequences of the C>T substitution at this SNP were assessed by cell-based reporter assays using three different cell lines. We found a repressive effect of the rs12203592 wildtype allele C on IRF4 promoter activity (P<0.001) but no repression by the variant allele in any cell line tested. Thus, homozygosity for the rs12203592 variant allele would result in increased IRF4 expression. This increase would be compounded by high levels of NF-kappaB activity in males due to the absence of estrogen. IRF4 differs from other IRFs in its anti-interferon activity which interferes with immune surveillance. We propose that a detailed study of IRF4 can provide information on the mechanism of the sex effect and the role of immune surveillance in childhood ALL development.
Collapse
Affiliation(s)
- Thuy N Do
- Genomic Immunoepidemiology Laboratory, HUMIGEN LLC, The Institute for Genetic Immunology, 2439 Kuser Road, Hamilton, NJ 08690-3303, USA
| | | | | | | | | |
Collapse
|
49
|
Wei CW, Lin CC, Yu YL, Lin CY, Lin PC, Wu MT, Chen CJ, Chang W, Lin SZ, Chen YLS, Harn HJ. n-Butylidenephthalide induced apoptosis in the A549 human lung adenocarcinoma cell line by coupled down-regulation of AP-2alpha and telomerase activity. Acta Pharmacol Sin 2009; 30:1297-306. [PMID: 19701232 DOI: 10.1038/aps.2009.124] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIM To investigate the role of hTERT gene expression and AP-2alpha in n-butylidenephthalide (n-BP)-induced apoptosis in A549 lung cancer cells. METHODS Viability of A549 cells was measured by MTT assay. Protein expression was determined by Western blot. Telomerase activity was measured using the modified telomere repeat amplification protocol (TRAP) assay. Xenograft mice were used as a model system to study the cytotoxic effect of n-BP in vivo. The morphology of tumor was examined by immunohistochemical staining. RESULTS The growth of A549 lung cancer cells treated with n-BP was significantly inhibited. Telomerase activity and hTERT mRNA expression were determined by telomeric repeat amplification protocol and reverse transcription-polymerase chain reaction, respectively. n-BP inhibited telomerase activity and hTERT mRNA expression in A549 cells while overexpression of hTERT could abolish BP-induced growth inhibition in the A549 cells. We also showed that hTERT promoter activity in the presence of n-BP was mediated via AP-2alpha. We saw an inhibition of tumor growth when nude mice carrying A549 subcutaneous xenograft tumors were treated with n-BP. Immunohistochemistry of this tumor tissue also showed a decrease in the expression of hTERT. CONCLUSION The antiproliferative effects of n-BP on A549 cells in vitro and in vivo suggest a novel clinical application of this compound in the treatment of lung cancers.
Collapse
|
50
|
Kerschgens J, Egener-Kuhn T, Mermod N. Protein-binding microarrays: probing disease markers at the interface of proteomics and genomics. Trends Mol Med 2009; 15:352-8. [DOI: 10.1016/j.molmed.2009.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 06/08/2009] [Accepted: 06/08/2009] [Indexed: 12/31/2022]
|