622 results on '"Proto-Oncogenes"'
Search Results
2. Canadian ROS proto-oncogene 1 study (CROS) for multi-institutional implementation of ROS1 testing in non-small cell lung cancer
- Author
-
Tracy Tucker, Margaret M. Kelly, Jean-Claude Cutz, Ming-Sound Tsao, Ranjit Waghray, Weei-Yuan Huang, Doha Itani, Christian Couture, Adam C. Smith, Carol C. Cheung, Fariboz Rashid-Kolvear, Emina Torlakovic, Yasushi Yatabe, Anna Bojarski, Michel R. Nasr, Aly Karsan, Zhaolin Xu, Gefei Qing, Iyare Izevbaye, Roula Albadine, Gilbert Bigras, Harmanjatinder S. Sekhon, Joan H.M. Knoll, Alan Spatz, H. Wang, Keith Kwan, Diana N. Ionescu, Tracy Stockley, and Danh Tran-Thanh
- Subjects
Pulmonary and Respiratory Medicine ,Canada ,Cancer Research ,Lung Neoplasms ,Proto-Oncogene Mas ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,Proto-Oncogenes ,ROS1 ,Humans ,Medicine ,Lung cancer ,In Situ Hybridization, Fluorescence ,Cellular localization ,Crizotinib ,Oncogene ,medicine.diagnostic_test ,biology ,business.industry ,Protein-Tyrosine Kinases ,medicine.disease ,Oncology ,Cancer research ,biology.protein ,Immunohistochemistry ,Antibody ,Reactive Oxygen Species ,business ,medicine.drug ,Fluorescence in situ hybridization - Abstract
Patients with non-small cell lung cancer (NSCLC) harboring ROS proto-oncogene 1 (ROS1) gene rearrangements show dramatic response to the tyrosine kinase inhibitor (TKI) crizotinib. Current best practice guidelines recommend that all advanced stage non-squamous NSCLC patients be also tested for ROS1 gene rearrangements. Several studies have suggested that ROS1 immunohistochemistry (IHC) using the D4D6 antibody may be used to screen for ROS1 fusion positive lung cancers, with assays showing high sensitivity but moderate to high specificity. A break apart fluorescence in situ hybridization (FISH) test is then used to confirm the presence of ROS1 gene rearrangement. The goal of Canadian ROS1 (CROS) study was to harmonize ROS1 laboratory developed testing (LDT) by using IHC and FISH assays to detect ROS1 rearranged lung cancers across Canadian pathology laboratories. Cell lines expressing different levels of ROS1 (high, low, none) were used to calibrate IHC protocols after which participating laboratories ran the calibrated protocols on a reference set of 24 NSCLC cases (9 ROS1 rearranged tumors and 15 ROS1 non-rearranged tumors as determined by FISH). Results were compared using a centralized readout. The stained slides were evaluated for the cellular localization of staining, intensity of staining, the presence of staining in non-tumor cells, the presence of non-specific staining (e.g. necrosis, extracellular mater, other) and the percent positive cells. H-score was also determined for each tumor. Analytical sensitivity and specificity harmonization was achieved by using low limit of detection (LOD) as either any positivity in the U118 cell line or H-score of 200 with the HCC78 cell line. An overall diagnostic sensitivity and specificity of up to 100% and 99% respectively was achieved for ROS1 IHC testing (relative to FISH) using an adjusted H-score readout on the reference cases. This study confirms that LDT ROS1 IHC assays can be highly sensitive and specific for detection of ROS1 rearrangements in NSCLC. As NSCLC can demonstrate ROS1 IHC positivity in FISH-negative cases, the degree of the specificity of the IHC assay, especially in highly sensitive protocols, is mostly dependent on the readout cut-off threshold. As ROS1 IHC is a screening assay for a rare rearrangements in NSCLC, we recommend adjustment of the readout threshold in order to balance specificity, rather than decreasing the overall analytical and diagnostic sensitivity of the protocols.
- Published
- 2021
3. Exon Definition Facilitates Reliable Control of Alternative Splicing in the RON Proto-Oncogene
- Author
-
Mihaela Enculescu, Julian Koenig, Samarth Thonta Setty, Stefan Legewie, Anke Busch, Simon Braun, and Kathi Zarnack
- Subjects
0303 health sciences ,Spliceosome ,Alternative splicing ,Biophysics ,Intron ,RNA ,Exons ,Computational biology ,Biology ,Article ,Introns ,Alternative Splicing ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Proto-Oncogenes ,Gene expression ,RNA splicing ,Glucans ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Alternative splicing is a key step in eukaryotic gene expression that allows for the production of multiple transcript and protein isoforms from the same gene. Even though splicing is perturbed in many diseases, we currently lack insights into regulatory mechanisms promoting its precision and efficiency. We analyze high-throughput mutagenesis data obtained for an alternatively spliced exon in the proto-oncogene RON and determine the functional units that control this splicing event. Using mathematical modeling of distinct splicing mechanisms, we show that alternative splicing is based in RON on a so-called “exon definition” mechanism. Here, the recognition of the adjacent exons by the spliceosome is required for removal of an intron. We use our model to analyze the differences between the exon and intron definition scenarios and find that exon definition prevents the accumulation of deleterious, partially spliced retention products during alternative splicing regulation. Furthermore, it modularizes splicing control, as multiple regulatory inputs are integrated into a common net input, irrespective of the location and nature of the corresponding cis-regulatory elements in the pre-messenger RNA. Our analysis suggests that exon definition promotes robust and reliable splicing outcomes in RON splicing.
- Published
- 2020
4. Upregulation of proto-oncogene ski by thyroid hormone in the intestine and tail during Xenopus metamorphosis
- Author
-
Liezhen Fu, Robert Liu, Vincent Ma, and Yun-Bo Shi
- Subjects
Thyroid Hormones ,Receptors, Thyroid Hormone ,Xenopus ,Metamorphosis, Biological ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Up-Regulation ,Intestines ,Xenopus laevis ,Retinoid X Receptors ,Endocrinology ,Larva ,Proto-Oncogenes ,Animals ,Triiodothyronine ,Animal Science and Zoology - Abstract
Thyroid hormone (T3) is important for adult organ function and vertebrate development, particularly during the postembryonic period when many organs develop/mature into their adult forms. Amphibian metamorphosis is totally dependent on T3 and can be easily manipulated, thus offering a unique opportunity for studying how T3 controls postembryonic development in vertebrates. Numerous early studies have demonstrated that T3 affects frog metamorphosis through T3 receptor (TR)-mediated regulation of T3 response genes, where TR forms a heterodimer with RXR (9-cis retinoic acid receptor) and binds to T3 response elements (TREs) in T3 response genes to regulate their expression. We have previously identified many candidate direct T3 response genes in Xenopus tropicalis tadpole intestine. Among them is the proto-oncogene Ski, which encodes a nuclear protein with complex function in regulating cell fate. We show here that Ski is upregulated in the intestine and tail of premetamorphic tadpoles upon T3 treatment and its expression peaks at stage 62, the climax of metamorphosis. We have further discovered a putative TRE in the first exon that can bind to TR/RXR in vitro and mediate T3 regulation of the promoter in vivo. These data demonstrate that Ski is activated by T3 through TR binding to a TRE in the first exon during Xenopus tropicalis metamorphosis, implicating a role of Ski in regulating cell fate during metamorphosis.
- Published
- 2022
5. Proto-oncogene FAM83A contributes to casein kinase 1–mediated mitochondrial maintenance and white adipocyte differentiation
- Author
-
Kuilong, Huang, Zhihao, Jia, Haoran, Li, Ying, Peng, Xiaochang, Chen, Nanjian, Luo, Tongxing, Song, Yingqian, Wang, Xin'e, Shi, Shihuan, Kuang, and Gongshe, Yang
- Subjects
Mice ,Adipogenesis ,Casein Kinase I ,3T3-L1 Cells ,Adipocytes, White ,Proto-Oncogenes ,Animals ,Cell Differentiation ,Cell Biology ,Molecular Biology ,Biochemistry ,Mitochondria ,Neoplasm Proteins - Abstract
Family with sequence similarity 83 A (FAM83A) is a newly discovered proto-oncogene that has been shown to play key roles in various cancers. However, the function of FAM83A in other physiological processes is not well known. Here, we report a novel function of FAM83A in adipocyte differentiation. We used an adipocyte-targeting fusion oligopeptide (FITC-ATS-9R) to deliver a FAM83A-sgRNA/Cas9 plasmid to knockdown Fam83a (ATS/sg-FAM83A) in white adipose tissue in mice, which resulted in reduced white adipose tissue mass, smaller adipocytes, and mitochondrial damage that was aggravated by a high-fat diet. In cultured 3T3-L1 adipocytes, we found loss or knockdown of Fam83a significantly repressed lipid droplet formation and downregulated the expression of lipogenic genes and proteins. Furthermore, inhibition of Fam83a decreased mitochondrial ATP production through blockage of the electron transport chain, associated with enhanced apoptosis. Mechanistically, we demonstrate FAM83A interacts with casein kinase 1 (CK1) and promotes the permeability of the mitochondrial outer membrane. Furthermore, loss of Fam83a in adipocytes hampered the formation of the TOM40 complex and impeded CK1-driven lipogenesis. Taken together, these results establish FAM83A as a critical regulator of mitochondria maintenance during adipogenesis.
- Published
- 2022
6. Applications of CRISPR/Cas9 Technology in the Treatment of Lung Cancer
- Author
-
Xiaohui Lin, Chunyang Jiang, and Zhigang Zhao
- Subjects
0301 basic medicine ,Lung Neoplasms ,Future studies ,T-Lymphocytes ,Genetic enhancement ,Programmed Cell Death 1 Receptor ,Drug Resistance ,Treatment of lung cancer ,Computational biology ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Mutation Rate ,Genome editing ,CRISPR-Associated Protein 9 ,Proto-Oncogenes ,medicine ,Animals ,Humans ,CRISPR ,Genes, Tumor Suppressor ,Gene Silencing ,Molecular Targeted Therapy ,Precision Medicine ,Lung cancer ,Molecular Biology ,Gene Editing ,business.industry ,Cas9 ,Genetic Therapy ,Oncogenes ,medicine.disease ,ErbB Receptors ,MicroRNAs ,Animal tumor ,030104 developmental biology ,Models, Animal ,Molecular Medicine ,gamma Catenin ,CRISPR-Cas Systems ,business ,030217 neurology & neurosurgery - Abstract
Since its emergence, the application of CRISPR-associated nuclease 9 (Cas9) technology in cancer research has accelerated studies to investigate many aspects of treatment approaches for lung cancer, including the identification of target genes, construction of animal tumor models, and identification of drug resistance-related genes. Moreover, CRISPR/Cas9 can be used in gene therapy for lung cancer, specifically involving molecular targeted drugs and inhibitors. This article reviews the current landscape of CRISPR/Cas9 applications for lung cancer treatment as a basis for further studies. Given its promising performance, in-depth and systematic research on the application of CRISPR/Cas9 in lung cancer treatment will be necessary in future studies for its successful implementation in clinical practice.
- Published
- 2019
7. The neural ELAVL protein HuB enhances endogenous proto-oncogene activation
- Author
-
Motoaki Yasuda, Fumihiro Higashino, Tomoyuki Hatanaka, and Kanchu Tei
- Subjects
Transcriptional Activation ,0301 basic medicine ,Proto-oncogene ,Carcinogenesis ,3 ' UTR ,Biophysics ,ELAV-Like Protein 2 ,Biology ,CREB ,Proto-Oncogene Mas ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Neoplasms ,Proto-Oncogenes ,Humans ,RNA, Messenger ,Nuclear export signal ,3' Untranslated Regions ,Molecular Biology ,Three prime untranslated region ,HuB ,Cell Biology ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cytosol ,030104 developmental biology ,Cytoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,HuR ,Ectopic expression - Abstract
The cytoplasmic distribution of the HuR/ELAVL1 (embryonic lethal abnormal vision 1) protein is recognized as an important prognostic factor of malignant tumors. However, the previous study suggests that exogenous over-expression of HuR is not sufficient for nuclear export. Conversely, the predominantly cytosolic distribution of neuron-specific human ELAV members, including HuB/ELAVL2, HuC/ELAVL3, and HuD/ELAVL4, has been reported. In the present study, we demonstrated the expression of HuB in several types of cancer cells, but expression of HuC and HuD was not observed. In addition, our results indicated that HuR and HuB formed a complex in the cytosolic fraction of cancer cells via the RRM3 region. Ectopic expression of HuB was capable of initiating the cytosolic translocation of HuR from the nucleus to the cytosol. Furthermore, HuB-transduced cancer cells displayed significant nuclear export of HuR, with quantitative PCR experiments revealing the simultaneous upregulation of HIF-1 alpha, c-Fos, c-MYC, and Ets2 basal mRNA expression. Phorbol 12-myristate 13-acetate (PMA)-stimulated HuB-transduced cells demonstrated significantly enhanced activation of endogenous c-Fos and CREB dependent cascades. Finally, co-transfection of HuB with the El region of type 5 human adenovirus significantly enhanced El transformation activities but that of HuR with the El region did not. Collectively, our findings suggest that the neural Hu family protein HuB plays a major role in the activation of memory-related proto-oncogenes. (C) 2019 Elsevier Inc. All rights reserved.
- Published
- 2019
8. The Dual Roles of the Atypical Protein Kinase Cs in Cancer
- Author
-
Miguel Reina-Campos, Jorge Moscat, and Maria T. Diaz-Meco
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Isozyme ,Article ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,Neoplasms ,PD-L1 ,Proto-Oncogenes ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Protein kinase A ,Protein Kinase Inhibitors ,Hedgehog ,Protein Kinase C ,Kinase ,Tumor Suppressor Proteins ,Cell Polarity ,Cancer ,Epithelial Cells ,Cell Biology ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,Isoenzymes ,Disease Models, Animal ,Cell Transformation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,biology.protein ,Carcinogenesis ,Signal Transduction - Abstract
Atypical protein kinase C (aPKC) isozymes, PKCλ/ι and PKCζ, are now considered fundamental regulators of tumorigenesis. However, the specific separation of functions that determine their different roles in cancer is still being unraveled. Both aPKCs have pleiotropic context-dependent functions that can translate into tumor-promoter or -suppressive functions. Here, we review early and more recent literature to discuss how the different tumor types, and their microenvironments, might account for the selective signaling of each aPKC isotype. This is of clinical relevance because a better understanding of the roles of these kinases is essential for the design of new anti-cancer treatments.
- Published
- 2019
9. Cancer-Associated Mutations but No Cancer: Insights into the Early Steps of Carcinogenesis and Implications for Early Cancer Detection
- Author
-
Scott R. Kennedy, Yuezheng Zhang, and Rosa Ana Risques
- Subjects
0301 basic medicine ,Aging ,Cancer Research ,Carcinogenesis ,Somatic cell ,DNA Mutational Analysis ,Normal tissue ,Disease ,Cancer detection ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Proto-Oncogenes ,Biomarkers, Tumor ,medicine ,Humans ,Early Cancer Detection ,Early Detection of Cancer ,Tumor Suppressor Proteins ,Positive selection ,High-Throughput Nucleotide Sequencing ,Cancer ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research - Abstract
Cancer is a disease of aging fueled by the accumulation of somatic mutations. While mutations in tumors are well characterized, little is known about the early mutational processes that initiate tumorigenesis. Recent advances in next-generation sequencing (NGS) have enabled the detection of mutations in normal tissue, revealing an unanticipated high level of age-related somatic mutations affecting most individuals and tissues. Surprisingly, many of these mutations are similar to mutations commonly found in tumors, suggesting an ongoing process of positive selection and clonal expansion akin to what occurs in cancer, but within normal tissue. Here we discuss some of the most important biological and clinical implications of these novel findings, with a special focus on their impact for cancer detection and prediction.
- Published
- 2019
10. The significance of gene mutations across eight major cancer types
- Author
-
Arup Kumar Malakar, Prosenjit Paul, and Supriyo Chakraborty
- Subjects
0301 basic medicine ,Carcinogenesis ,Health, Toxicology and Mutagenesis ,Gene mutation ,Biology ,medicine.disease_cause ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Proto-Oncogenes ,Heredity ,Genetic variation ,Genetics ,medicine ,Animals ,Humans ,Cancer biology ,Gene ,Mutation ,Cancer ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis - Abstract
Mutations occur spontaneously, which can be induced by either chemicals (e.g. benzene) or biological factors (e.g. virus). Not all mutations cause noticeable changes in cellular functions. However, mutation in key cellular genes leads to developmental disorders. It is one of the main ways in which proto-oncogenes can be changed into their oncogenic state. The progressive accumulation of multiple mutations throughout life leads to cancer. In the past few decades, extensive research on cancer biology has discovered many genes and pathways having role in cancer development. In this review, we tried to summarize the current knowledge of mutational effect on different cancer types and its consequences in brief for future reference and guidance of researchers in cancer biology.
- Published
- 2019
11. Persistent Disease and Recurrence in Medullary Thyroid Carcinoma: A Case Series
- Author
-
Fructuoso Rodríguez Rodríguez, Juan Carlos Rocca Cardenas, Ana Alicia Tejera Hernández, Juan Ramón Hernández Hernández, and María Isabel Gutiérrez Giner
- Subjects
Adult ,Calcitonin ,Male ,medicine.medical_specialty ,Medullary cavity ,medicine.medical_treatment ,MEDLINE ,Thyroid carcinoma ,Postoperative Complications ,Proto-Oncogenes ,Carcinoma ,medicine ,Humans ,Thyroid Neoplasms ,Neoplasm Metastasis ,Survival analysis ,Aged ,Series (stratigraphy) ,business.industry ,General Engineering ,Middle Aged ,medicine.disease ,Survival Analysis ,Carcinoma, Neuroendocrine ,Radiation therapy ,Persistent Disease ,Thyroidectomy ,Female ,Radiotherapy, Adjuvant ,Lymph Nodes ,Radiology ,Neoplasm Recurrence, Local ,business - Published
- 2019
12. Context-dependent functions of KLF4 in cancers: Could alternative splicing isoforms be the key?
- Author
-
John R. Stroehlein, Liang Wang, Daoyan Wei, and Feng Shen
- Subjects
0301 basic medicine ,Gene isoform ,Cancer Research ,Kruppel-Like Transcription Factors ,Context (language use) ,Biology ,Kruppel-Like Factor 4 ,03 medical and health sciences ,0302 clinical medicine ,stomatognathic system ,Neoplasms ,Proto-Oncogenes ,Animals ,Humans ,Protein Isoforms ,Gene ,Transcription factor ,Tumor Suppressor Proteins ,fungi ,Alternative splicing ,Survival Analysis ,Cell biology ,Gene Expression Regulation, Neoplastic ,Alternative Splicing ,030104 developmental biology ,Oncology ,KLF4 ,030220 oncology & carcinogenesis ,embryonic structures ,sense organs ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Reprogramming - Abstract
Krüppel-like factor 4 (KLF4) is an important transcription factor that is expressed in a variety of tissues and regulates many critical physiologic and cellular processes, including cell proliferation, differentiation, stem cell reprogramming, maintenance of genomic stability, and normal tissue homeostasis. KLF4 has both tumor suppressive and oncogenic functions in gastrointestinal and other cancers. These functions are thought to be context dependent, but how KLF4 exerts these differential functions and the molecular mechanisms behind them remain poorly understood. Recent studies have shown that the KLF4 gene undergoes alternative splicing, and the protein products of certain transcripts antagonize wild-type KLF4 function, suggesting an additional layer of regulation of KLF4 function. Therefore, detailed study of KLF4 alternative splicing may not only provide new insights into the complexity of KLF4 functions but also lead to rational targeting of KLF4 for cancer prevention and therapy.
- Published
- 2018
13. microRNA-181a-5p impedes the proliferation, migration, and invasion of retinoblastoma cells by targeting the NRAS proto-oncogene
- Author
-
Ming Ouyang, Guiqin Liu, Cheng Xiong, and Jing Rao
- Subjects
Retinal Neoplasms ,miR-181a-5p ,Retinoblastoma ,Membrane Proteins ,NRAS ,General Medicine ,GTP Phosphohydrolases ,MicroRNAs ,Cell Movement ,Cell Line, Tumor ,Proto-Oncogenes ,Humans ,Neoplasm Invasiveness ,Cell Proliferation - Abstract
Objectives Accumulating research have reported that microRNAs (miRNAs) play important roles in Retinoblastoma (RB). Nonetheless, the function and underlying mechanism of miR-181a-5p in RB remain ambiguous. Methods The relative expression levels of miR-181a-5p and NRAS mRNA were detected by quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR). RB cell proliferation was measured using the Cell Counting Kit-8 (CCK-8) and 5′-Bromo-2′-deoxyuridine (BrdU) assays. Transwell assays and flow cytometry were performed to detect the migration, invasion, and apoptosis of RB cells. The interaction between miR-181a-5p and NRAS was explored using luciferase experiments, western blotting, and qRT-PCR. Results miR-181a-5p expression was found to be decreased in RB tissues and cell lines, and its expression was correlated with unfavorable pathological features of the patients. In vitro experiments revealed that miR-181a-5p reduced RB cell proliferation, migration, and invasion while enhancing apoptosis. Further research confirmed that NRAS is a direct target of miR-181a-5p. miR-181a-5p inhibited NRAS expression at both the mRNA and protein levels. Co-transfection of pcDNA-NRAS or NRAS small interfering RNA (siRNA) reversed the effects of miR-181a-5p mimics or miR-181a-5p inhibitors on RB cells. Conclusion miR-181a-5p was significantly downregulated during the development of RB, and it suppressed the malignant behaviors of RB cells by targeting NRAS.
- Published
- 2022
14. Angiotensin receptor blockade attenuates cholangiocarcinoma cell growth by inhibiting the oncogenic activity of Yes-associated protein
- Author
-
Keisuke Nakanishi, Akira Mitoro, Mitsuteru Kitade, Soichiro Saikawa, Koh Kitagawa, Norihisa Nishimura, Shinya Sato, Hideto Kawaratani, Tadashi Namisaki, Kei Moriya, Kenichiro Seki, Hitoshi Yoshiji, and Kosuke Kaji
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Angiotensin receptor ,Angiogenesis ,Mice, Nude ,Losartan ,Cholangiocarcinoma ,Angiotensin Receptor Antagonists ,03 medical and health sciences ,Cell Line, Tumor ,Proto-Oncogenes ,parasitic diseases ,Animals ,Humans ,Medicine ,cardiovascular diseases ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Mice, Inbred BALB C ,Oncogene ,business.industry ,Cell growth ,YAP-Signaling Proteins ,Phosphoproteins ,Xenograft Model Antitumor Assays ,Angiotensin II ,Gene Expression Regulation, Neoplastic ,CTGF ,030104 developmental biology ,Bile Duct Neoplasms ,Oncology ,CYR61 ,cardiovascular system ,Cancer research ,business ,Transcription Factors ,medicine.drug - Abstract
Cholangiocarcinoma (CCA) is a destructive malignancy with limited responsiveness to conventional chemotherapy. Although angiotensin receptor blockers (ARBs) have gained attention for their potential anticancer activity, little is known about their effects on CCA. The transcriptional co-activator, Yes-associated protein (YAP) is a critical oncogene in several cancers, including CCA. Following recent evidence showing that YAP is regulated by angiotensin II (AT-II), we investigated the effects of an ARB, losartan, on two human CCA cell lines (KKU-M213 and HuCCT-1) with regards to YAP oncogenic regulation. Losartan suppressed AT-II-induced CCA cell proliferation in a dose-dependent manner, induced apoptosis, decreased YAP (Ser127), and downregulated the YAP target genes CTGF, CYR61, ANKRD1, and MFAP5. However, losartan did not affect epithelial-mesenchymal transition, differentiation, or stemness in the CCA cells. Xenograft tumor growth assay showed that oral administration of a low clinical dose of losartan considerably reduced subcutaneous tumor burden and attenuated intratumor vascularization in CCA cell-derived xenograft tumors in BALB/c nude mice. These results indicate that ARB therapy could serve as a potential novel strategy for CCA treatment.
- Published
- 2018
15. Pericardial and Pleural Metastases: Clinical, Histologic, and Molecular Differences
- Author
-
Georgia Karpathiou, Michel Peoc'h, Mousa Mobarki, Arnaud Patoir, Marie Laure Stachowicz, Marios Froudarakis, Sirine Hathroubi, and Olivier Tiffet
- Subjects
Male ,Pathology ,Lung Neoplasms ,Pleural effusion ,medicine.disease_cause ,Metastasis ,Cohort Studies ,0302 clinical medicine ,Medicine ,Pericardium ,medicine.diagnostic_test ,Biopsy, Needle ,Middle Aged ,Prognosis ,Immunohistochemistry ,Lymphatic system ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,KRAS ,Cardiology and Cardiovascular Medicine ,Adult ,Proto-Oncogene Proteins B-raf ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Adenocarcinoma ,Risk Assessment ,Sensitivity and Specificity ,Pericardial Effusion ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Proto-Oncogenes ,Biopsy ,Biomarkers, Tumor ,Humans ,Neoplasm Invasiveness ,Aged ,Neoplasm Staging ,Retrospective Studies ,Lung ,business.industry ,Genes, erbB-1 ,medicine.disease ,Survival Analysis ,Pleural Effusion, Malignant ,respiratory tract diseases ,030228 respiratory system ,Surgery ,business - Abstract
Pericardial and pleural cavities produce effusions with important clinical consequences. Metastases are one of the most common etiologies of both serosal effusions. However, data regarding the type of metastatic involvement of the pleura and the pericardium are lacking. This study investigated the histologic patterns of tumors involving the two cavities to better define their pathophysiology and possible consequences in molecular diagnostics.This was a retrospective study of patients diagnosed with pericardial (n = 75) and pleural (n = 70) metastases. Patterns of metastasis were characterized as (1) tumor cells floating inside the cavity (2) as lymphatic emboli and (3) as tumor cells frankly invading underlying fibrous tissue. Molecular analysis (EGFR, KRAS, BRAF, ALK, HER2) was performed in 44 metastases of lung adenocarcinomas.The two serosal membranes differed significantly (p0.0001) in the pattern of metastasis. The pleura showed predominantly an invasive pattern (67 [95.7%]), whereas most pericardial metastases consisted of tumor cells floating inside the cavity or as lymphatic emboli (44 [58.6%]). The origin of the primary differed marginally between the two organs. Time to diagnosis of metastasis differed between the two organs, with pleural metastases presenting later than the pericardial ones. Molecular analysis failed more often in pericardial biopsy specimens and in specimens with emboli or surface involvement.Although pericardium and pleura share common embryologic and histologic features and are often regarded as giving similar effusions, they differ significantly in the type of metastases involving them. This can have important consequences in histologic, cytologic, and molecular diagnostics.
- Published
- 2018
16. The MDS and EVI1 complex locus (MECOM) isoforms regulate their own transcription and have different roles in the transformation of hematopoietic stem and progenitor cells
- Author
-
María A. García-Sánchez, Rafael Alis, Nerea Marcotegui, Leire Urquiza, Ion Cristóbal, Miren Maicas, Iria Vázquez, Xabier Cortes-Lavaud, and María D. Odero
- Subjects
0301 basic medicine ,Gene isoform ,MECOM ,Response element ,Biophysics ,Biology ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Structural Biology ,Transcription (biology) ,Proto-Oncogenes ,Genetics ,Animals ,Humans ,Progenitor cell ,Promoter Regions, Genetic ,Molecular Biology ,Transcription factor ,Leukemia ,Gene Expression Regulation, Leukemic ,Promoter ,Hematopoietic Stem Cells ,MDS1 and EVI1 Complex Locus Protein ,Cell biology ,DNA-Binding Proteins ,Cell Transformation, Neoplastic ,030104 developmental biology ,RUNX1 ,chemistry ,Transcription Factors - Abstract
Transcriptional activation of the EVI1 oncogene (3q26) leads to aggressive forms of human acute myeloid leukemia (AML). However, the mechanism of EVI1-mediated leukemogenesis has not been fully elucidated. Previously, by characterizing the EVI1 promoter, we have shown that RUNX1 and ELK1 directly regulate EVI1 transcription. Intriguingly, bioinformatic analysis of the EVI1 promoter region identified the presence of several EVI1 potential binding sites. Thus, we hypothesized that EVI1 could bind to these sites regulating its own transcription. In this study, we show that there is a functional interaction between EVI1 and its promoter, and that the different EVI1 isoforms (EVI1-145kDa, EVI1-Δ324 and MDS1-EVI1) regulate the transcription of EVI1 transcripts through distinct promoter regions. Moreover, we determine that the EVI1-145kDa isoform activates EVI1 transcription, whereas EVI1-Δ324 and MDS1-EVI1 act as repressors. Finally, we demonstrate that these EVI1 isoforms are involved in cell transformation; functional experiments show that EVI1-145kDa prolongs the maintenance of hematopoietic stem and progenitor cells; conversely, MDS1-EVI1 repressed hematopoietic stem and progenitor colony replating capacity. We demonstrate for the first time that EVI1 acts as a regulator of its own expression, highlighting the complex regulation of EVI1, and open new directions to better understand the mechanisms of EVI1 overexpressing leukemias.
- Published
- 2017
17. Activation of EVI1 transcription by the LEF1/β-catenin complex with p53-alteration in myeloid blast crisis of chronic myeloid leukemia
- Author
-
Motomi Osato, Nawin Manachai, Avinash Govind Bahirvani, Yusuke Saito, Kazuhiro Morishita, and Shingo Nakahata
- Subjects
Transcriptional Activation ,0301 basic medicine ,Myeloid ,Lymphoid Enhancer-Binding Factor 1 ,Biophysics ,Biology ,Biochemistry ,Fusion gene ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Proto-Oncogenes ,Transcriptional regulation ,medicine ,Animals ,Humans ,neoplasms ,Molecular Biology ,beta Catenin ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,Imatinib ,Cell Biology ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,DNA-Binding Proteins ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Phosphorylation ,Catenin complex ,Tumor Suppressor Protein p53 ,Blast Crisis ,Transcription Factors ,medicine.drug - Abstract
The presence of a BCR-ABL1 fusion gene is necessary for the pathogenesis of chronic myeloid leukemia (CML) through t(9;22)(q34;q11) translocation. Imatinib, an ABL tyrosine kinase inhibitor, is dramatically effective in CML patients; however, 30% of CML patients will need further treatment due to progression of CML to blast crisis (BC). Aberrant high expression of ecotropic viral integration site 1 (EVI1) is frequently observed in CML during myeloid-BC as a potent driver with a CML stem cell signature; however, the precise molecular mechanism of EVI1 transcriptional regulation during CML progression is poorly defined. Here, we demonstrate the transcriptional activity of EVI1 is dependent on activation of lymphoid enhancer-binding factor 1 (LEF1)/β-catenin complex by BCR-ABL with loss of p53 function during CML-BC. The activation of β-catenin is partly dependent on BCR-ABL expression through enhanced GSK3β phosphorylation, and EVI1 expression is directly enhanced by the LEF1/β-catenin complex bound to the EVI1 promoter region. Moreover, the loss of p53 expression is inversely correlated with high expression of EVI1 in CML leukemia cells with an aggressive phase of CML, and a portion of the activation mechanism of EVI1 expression is dependent on β-catenin activation through GSK3β phosphorylation by loss of p53. Therefore, we found that the EVI1 activation in CML-BC is dependent on LEF1/β-catenin activation by BCR-ABL expression with loss of p53 function, representing a novel selective therapeutic approach targeting myeloid blast crisis progression.
- Published
- 2017
18. MicroRNA-24 increases hepatocellular carcinoma cell metastasis and invasion by targeting p53: miR-24 targeted p53
- Author
-
Li Chen, Fan Chen, Lian-zhou Chen, Wei Chen, Jing-song Chen, Hong-Xu Xu, Wen-Tao Zeng, Liang Luo, and Xiao-Hui Huang
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Down-Regulation ,Biology ,Transfection ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,RNA interference ,Cell Line, Tumor ,Internal medicine ,Proto-Oncogenes ,microRNA ,medicine ,Humans ,Neoplasm Invasiveness ,3' Untranslated Regions ,Pharmacology ,Gene knockdown ,Binding Sites ,Oncogene ,Three prime untranslated region ,Liver Neoplasms ,General Medicine ,medicine.disease ,digestive system diseases ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,RNA Interference ,Tumor Suppressor Protein p53 ,Signal Transduction - Abstract
MicroRNA-24 (miR-24), a member of the miRNA family, functions as an oncogene in various types of human cancer. However, the underlying mechanisms of miR-24 involvement in the development and progression of hepatocellular carcinoma (HCC) remain poorly understood. The present study revealed that miRNA-24 down-regulates p53 through binding to the 3'-UTR of p53 mRNA based on a luciferase reporter assay, and that the expression level of miR-24 could affect the invasion of HCC lines via p53. Down-regulation of p53 significantly attenuated the inhibitory effects of miR-24 knockdown on the invasion of HCC cells, suggesting that miR-24 could be a potential target for HCC treatment. Moreover, our results revealed that miR-24 expression was significantly increased in HCC metastatic tumor tissues compared with matched non-metastatic tumor tissues, and that the up-regulation of miR-24 was significantly associated with down-regulation of p53 in the HCC tissues. In conclusion, this study demonstrates that miR-24 functions as an oncogene in HCC, at least partly by promoting cell invasion through down-regulation of p53. Therefore, miR-24 may be a potential therapeutic target for treatment of HCC.
- Published
- 2016
19. Cooperation Between Distinct Cancer Driver Genes Underlies Intertumor Heterogeneity in Hepatocellular Carcinoma
- Author
-
Pedro Molina-Sánchez, Kai Nie, Tiphaine Martin, Ying Ding, Elizabeth Bitterman, Robert A. Rollins, Troy Rubenstein, Abdulkadir Elmas, Erin Bresnahan, Marina Bárcena-Varela, Veronica Miguela, Melissa Yao, David J. Shields, Marina Ruiz de Galarreta, Katherine E. Lindblad, Jonathan Golas, Lauren Tal Grinspan, Ramon Parsons, Shambhunath Choudhary, Zhengyan Kan, Kuan-Lin Huang, and Amaia Lujambio
- Subjects
Male ,0301 basic medicine ,Carcinoma, Hepatocellular ,Mice, Transgenic ,Biology ,Article ,Transcriptome ,Genetic Heterogeneity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Proto-Oncogenes ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,neoplasms ,Gene ,PI3K/AKT/mTOR pathway ,Hepatology ,Oncogene ,Liver Neoplasms ,Gastroenterology ,Computational Biology ,Cancer ,HCCS ,medicine.disease ,Phenotype ,digestive system diseases ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,Female ,Tumor Escape ,030211 gastroenterology & hepatology - Abstract
Background And Aims The pattern of genetic alterations in cancer driver genes in patients with hepatocellular carcinoma (HCC) is highly diverse, which partially explains the low efficacy of available therapies. In spite of this, the existing mouse models only recapitulate a small portion of HCC inter-tumor heterogeneity, limiting the understanding of the disease and the nomination of personalized therapies. Here, we aimed at establishing a novel collection of HCC mouse models that captured human HCC diversity. Methods By performing hydrodynamic tail-vein injections, we tested the impact of altering a well-established HCC oncogene (either MYC or β-catenin) in combination with an additional alteration in one of eleven other genes frequently mutated in HCC. Of the 23 unique pairs of genetic alterations that we interrogated, 9 were able to induce HCC. The established HCC mouse models were characterized at histopathological, immune, and transcriptomic level to identify the unique features of each model. Murine HCC cell lines were generated from each tumor model, characterized transcriptionally, and used to identify specific therapies that were validated in vivo. Results Cooperation between pairs of driver genes produced HCCs with diverse histopathology, immune microenvironments, transcriptomes, and drug responses. Interestingly, MYC expression levels strongly influenced β-catenin activity, indicating that inter-tumor heterogeneity emerges not only from specific combinations of genetic alterations but also from the acquisition of expression-dependent phenotypes. Conclusions This novel collection of murine HCC models and corresponding cell lines establishes the role of driver genes in diverse contexts and enables mechanistic and translational studies.
- Published
- 2020
20. EVI1 Interferes with Myeloid Maturation via Transcriptional Repression of Cebpa, via Binding to Two Far Downstream Regulatory Elements
- Author
-
Ying-Yi Xiao, Archibald S. Perkins, Yi Zhang, Minh Tran, Vasiliki Tsakraklides, and Michael P Wilson
- Subjects
0301 basic medicine ,Myeloid ,Transcription, Genetic ,Cellular differentiation ,Bone Marrow Cells ,Biology ,Response Elements ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cricetinae ,Proto-Oncogenes ,CEBPA ,medicine ,Animals ,Antigens, Ly ,Progenitor cell ,Molecular Biology ,Transcription factor ,Leukemia ,Membrane Proteins ,Myeloid leukemia ,Molecular Bases of Disease ,Cell Differentiation ,Cell Biology ,Hematopoietic Stem Cells ,medicine.disease ,Molecular biology ,MDS1 and EVI1 Complex Locus Protein ,Cell biology ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,CCAAT-Enhancer-Binding Proteins ,Stem cell ,Transcription Factors - Abstract
One mechanism by which oncoproteins work is through perturbation of cellular maturation; understanding the mechanisms by which this occurs can lead to the development of targeted therapies. EVI1 is a zinc finger oncoprotein involved in the development of acute myeloid leukemia; previous work has shown it to interfere with the maturation of granulocytes from immature precursors. Here we investigate the mechanism by which that occurs, using an immortalized hematopoietic progenitor cell line, EML-C1, as a model system. We document that overexpression of EVI1 abrogates retinoic acid-induced maturation of EML cells into committed myeloid cells, a process that can be documented by the down-regulation of stem cell antigen-1 and acquisition of responsiveness to granulocyte-macrophage colony-stimulating factor. We show that this requires DNA binding capacity of EVI1, suggesting that downstream target genes are involved. We identify the myeloid regulator Cebpa as a target gene and identify two EVI1 binding regions within evolutionarily conserved enhancer elements at +35 and +37 kb relative to the gene. EVI1 can strongly suppress Cebpa transcription, and add-back of Cebpa into EVI1-expressing EML cells partially corrects the block in maturation. We identify the DNA sequences to which EVI1 binds at +35 and +37 kb and show that mutation of one of these releases Cebpa from EVI1-induced suppression. We observe a more complex picture in primary bone marrow cells, where EVI1 suppresses Cebpa in stem cells but not in more committed progenitors. Our data thus identify a regulatory node by which EVI1 contributes to leukemia, and this represents a possible therapeutic target for treatment of EVI1-expressing leukemia.
- Published
- 2016
21. Identification of the specific epigenetic alterations associated with chemo-resistance via reprogramming of cancer cells
- Author
-
Jong-Joo Kim and Rajani Rai
- Subjects
Genetics ,Proto-Oncogenes ,Somatic cell ,Cancer ,Antineoplastic Agents ,General Medicine ,Biology ,Cellular Reprogramming ,medicine.disease ,MLH1 ,Adaptation, Physiological ,Models, Biological ,Phenotype ,Epigenesis, Genetic ,Neoplasm Proteins ,Drug Resistance, Neoplasm ,Neoplasms ,Cancer cell ,Cancer research ,medicine ,Animals ,Humans ,Epigenetics ,Reprogramming - Abstract
Background Chemo-resistance is the main obstacle in cancer therapy, limiting the effectiveness of drug treatment. Epigenetics-mediated changes are suggested as a critical factor paying the chemo-resistance phenotype. Since epigenetic modulations are a reversible phenomenon, reversion of epigenetic changes represents a promising therapeutic approach for cancer. However, heterogeneity in epigenetic marks in tumor cells makes it difficult to identify the specific epigenetic aberrations contributing to chemo-resistance. Our hypothesis aimed to explore this issue to add therapeutic options for cancer. Presentation of the hypothesis Epigenetic alterations, the main mediator of cellular reprogramming, occur rapidly upon exposure to chemotherapy. Recent studies have demonstrated that reprogramming resets/erases the epigenetic marks established during differentiation to specific somatic cell types. To overcome the heterogeneous nature of cancer cells, we will attempt to make homogenous cancer cell colonies by reprogramming. Comparison of the drug-resistant cancer cells obtained from these colonies to parent cancer cells and reprogrammed cancer cells is an effective way to determine the precise epigenetic alterations underlying specific chemo-resistance. Testing the hypothesis Cellular reprogramming of cancer cells led to generation of homogenous colonies. Following lineage specification and long term drug treatment, the obtained drug resistance cells will be compared with parent cancer cells for whole genome epigenetic signature. Implications of the hypothesis A key implication of this hypothesis is that determination of the usefulness of cellular reprogramming of cancer cells enabling the identification of specific epigenetic modulation associated with particular drug resistance will enable exploration of new research avenues for cancer treatment.
- Published
- 2015
22. Quantification of EVI1 transcript levels in acute myeloid leukemia by RT-qPCR analysis: A study by the ALFA Group
- Author
-
Karine Celli-Lebras, Nicolas Boissel, Aline Renneville, Xavier Thomas, Thomas Smol, Christine Terré, B. Quesnel, Alice Marceau-Renaut, Claude Preudhomme, Hervé Dombret, Sylvie Castaigne, Céline Berthon, and Olivier Nibourel
- Subjects
Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Neoplasm, Residual ,Adolescent ,Transcription, Genetic ,Bone Marrow Cells ,Biology ,Real-Time Polymerase Chain Reaction ,Sensitivity and Specificity ,Young Adult ,Exon ,Plasmid ,Proto-Oncogenes ,medicine ,Humans ,Multicenter Studies as Topic ,splice ,RNA, Messenger ,RNA, Neoplasm ,Aged ,Randomized Controlled Trials as Topic ,Blood Cells ,Gene Expression Regulation, Leukemic ,Alternative splicing ,Myeloid leukemia ,Hematology ,Middle Aged ,Minimal residual disease ,Molecular biology ,MDS1 and EVI1 Complex Locus Protein ,Neoplasm Proteins ,DNA-Binding Proteins ,Leukemia, Myeloid, Acute ,Real-time polymerase chain reaction ,medicine.anatomical_structure ,Oncology ,Female ,Bone marrow ,Follow-Up Studies ,Transcription Factors - Abstract
EVI1 overexpression confers poor prognosis in acute myeloid leukemia (AML). Quantification of EVI1 expression has been mainly assessed by real-time quantitative PCR (RT-qPCR) based on relative quantification of EVI1-1D splice variant. In this study, we developed a RT-qPCR assay to perform quantification of EVI1 expression covering the different splice variants. A sequence localized in EVI1 exons 14 and 15 was cloned into plasmids that were used to establish RT-qPCR standard curves. Threshold values to define EVI1 overexpression were determined using 17 bone marrow (BM) and 31 peripheral blood (PB) control samples and were set at 1% in BM and 0.5% in PB. Samples from 64 AML patients overexpressing EVI1 included in the ALFA-0701 or -0702 trials were collected at diagnosis and during follow-up (n=152). Median EVI1 expression at AML diagnosis was 23.3% in BM and 3.6% in PB. EVI1 expression levels significantly decreased between diagnostic and post-induction samples, with an average variation from 21.6% to 3.56% in BM and from 4.0% to 0.22% in PB, but did not exceed 1 log10 reduction. Our study demonstrates that the magnitude of reduction in EVI1 expression levels between AML diagnosis and follow-up is not sufficient to allow sensitive detection of minimal residual disease.
- Published
- 2015
23. Dietary zinc deficiency or supplementation during gestation increases breast cancer susceptibility in adult female mice offspring following a J-shaped pattern and through distinct mechanisms
- Author
-
Fábia de Oliveira Andrade, Thomas Prates Ong, Luis Fernando Barbisan, Raquel Santana da Cruz, Vivian Montes de Oca Carioni, Mariana Papaléo Rosim, Inar Alves de Castro, Mayara Lilian Paulino Miranda, Camile Castilho Fontelles, Pedro Vitoriano de Oliveira, Universidade de São Paulo (USP), Food Research Center (FoRC), and Universidade Estadual Paulista (Unesp)
- Subjects
Male ,NUTRIÇÃO PRÉ-NATAL ,medicine.medical_specialty ,Offspring ,chemistry.chemical_element ,Apoptosis ,Zinc ,Biology ,Toxicology ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Breast cancer ,0404 agricultural biotechnology ,Pregnancy ,Internal medicine ,Fetal programming ,Proto-Oncogenes ,medicine ,Animals ,Maternal nutrition ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Fetus ,Gene Expression Profiling ,Mammary Neoplasms, Experimental ,04 agricultural and veterinary sciences ,General Medicine ,Malondialdehyde ,medicine.disease ,040401 food science ,Mice, Inbred C57BL ,Endocrinology ,chemistry ,Dietary Supplements ,Zinc deficiency ,Gestation ,Female ,Tumor Suppressor Protein p53 ,Deficiency Diseases ,Food Science - Abstract
Made available in DSpace on 2019-10-06T16:47:50Z (GMT). No. of bitstreams: 0 Previous issue date: 2019-12-01 Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) Zinc is required for fetal development and is involved in key processes associated with breast carcinogenesis. We evaluated whether maternal zinc deficiency or supplementation during gestation influences female offspring susceptibility to breast cancer in adulthood. C57BL/6 mice consumed during gestation control (30 p.p.m. zinc), zinc-deficient (8 p.p.m) or zinc-supplemented (45 p.p.m.) diets. Maternal zinc supplementation increased in female mice offspring the incidence of chemically-induced mammary adenocarcinomas that were heavier, compared to control group. This was accompanied by a decreased number of terminal end buds, increased cell proliferation and apoptosis, and increased tumor suppressors p21, p53 and Rassf1, Zfp382 and Stat3 expression in mammary glands, as well as increased zinc status. Although maternal zinc deficiency did not alter the incidence of these lesions, it also induced heavier mammary adenocarcinomas, compared to control group. These effects were accompanied by a decreased number of terminal end buds, increased proto-oncogenes c-Myc and Lmo4 expression and H3K9Me3 and H4K20Me3 epigenetic marks in mammary glands of offspring, and decreased zinc status and increased levels of oxidative marker malondialdehyde. The data suggest that both maternal zinc deficiency and supplementation during gestation programmed increased breast cancer susceptibility in adult mice offspring following a J-shaped pattern through distinct mechanisms. Department of Food Science and Nutrition School of Pharmaceutical Sciences University of São Paulo (USP) Department of Fundamental Chemistry University of São Paulo (USP) Food Research Center (FoRC) Department of Morphology Institute of Biosciences of Botucatu Sao Paulo State University (UNESP), São Paulo Department of Morphology Institute of Biosciences of Botucatu Sao Paulo State University (UNESP), São Paulo FAPESP: 11/23259-4 FAPESP: 2013/04960-9 CNPq: 307910/2016-4
- Published
- 2019
24. MicroRNA-128b suppresses tumor growth and promotes apoptosis by targeting A2bR in gastric cancer
- Author
-
Wei Zong, Guisheng Liu, Ping Wang, Xueyan Guo, Bin Song, and Shuixiang He
- Subjects
Biophysics ,Apoptosis ,Biology ,Receptor, Adenosine A2B ,Proto-Oncogene Mas ,Biochemistry ,Cell Movement ,Reference Values ,Stomach Neoplasms ,Cell Line, Tumor ,Proto-Oncogenes ,microRNA ,medicine ,Humans ,Molecular Biology ,Gene ,Cell Proliferation ,Reporter gene ,Oncogene ,Cell growth ,Cancer ,Cell Biology ,medicine.disease ,Molecular biology ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Cell culture ,Cancer research - Abstract
MicroRNAs (miRNAs) play crucial roles in the development and progression of human cancers, including gastric cancer (GC). The discovery of miRNAs may provide a new and powerful tool for studying the mechanism, diagnosis, and treatment of GC. In this study, we aimed to investigate the role and mechanism of miR-128b in the development and progression of GC. Quantitative real-time PCR (qRT-PCR) was used to measure the expression level of miR-128b in GC tissues and cell lines. We found that miR-128b was significantly down-regulated in GC tissues and cell lines. In addition, over-expression of miR-128b inhibited GC cell proliferation, migration and invasion of GC cells in vitro. Gain-of-function in vitro experiments further showed that the miR-128b mimic significantly promoted GC cell apoptosis. Subsequent dual-luciferase reporter assay identified one of the proto-oncogene A2bR as direct target of miR-128b. Therefore, our results indicate that miR-128b is a proto-oncogene miRNA that can suppresses GC proliferation and migration through down-regulation of the oncogene gene A2bR. Taken together, our results indicate that miR-128b could serve as a potential diagnostic biomarker and therapeutic option for human GC in the near future.
- Published
- 2015
25. The role of EVI1 in myeloid malignancies
- Author
-
Ruby Gonzalez, Yi Zhang, Michael Wilson, Carolyn Glass, and Archibald S. Perkins
- Subjects
Myeloid ,Biology ,Epigenesis, Genetic ,Mice ,Proto-Oncogenes ,medicine ,Transcriptional regulation ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Chromosome Aberrations ,Zinc finger ,Oncogene ,Gene Expression Regulation, Leukemic ,RUNX1T1 ,Myeloid leukemia ,Cell Biology ,Hematology ,MDS1 and EVI1 Complex Locus Protein ,DNA-Binding Proteins ,Disease Models, Animal ,medicine.anatomical_structure ,Genetic Loci ,Leukemia, Myeloid ,Histone methyltransferase ,Immunology ,Cancer research ,Molecular Medicine ,Signal Transduction ,Transcription Factors - Abstract
The EVI1 oncogene at human chr 3q26 is rearranged and/or overexpressed in a subset of acute myeloid leukemias and myelodysplasias. The EVI1 protein is a 135 kDa transcriptional regulator with DNA-binding zinc finger domains. Here we provide a critical review of the current state of research into the molecular mechanisms by which this gene plays a role in myeloid malignancies. The major pertinent cellular effects are blocking myeloid differentiation and preventing cellular apoptosis, and several potential mechanisms for these phenomena have been identified. Evidence supports a role for EVI1 in inducing cellular quiescence, and this may contribute to the resistance to chemotherapy seen in patients with neoplasms that overexpress EVI1. Another isoform, MDS1-EVI1 (or PRDM3), encoded by the same locus as EVI1, harbors an N-terminal histone methyltransferase(HMT) domain; experimental findings indicate that this protein and its HMT activity are critical for the progression of a subset of AMLs, and this provides a potential target for therapeutic intervention.
- Published
- 2014
26. miR-223 Regulates Cell Growth and Targets Proto-Oncogenes in Mycosis Fungoides/Cutaneous T-Cell Lymphoma
- Author
-
John A. Zic, Jeffrey P. Zwerner, Laura Y. McGirt, Devin A. Baerenwald, Christine M. Eischen, and Clare M. Adams
- Subjects
Cell Survival ,Biopsy ,Cell ,Dermatology ,Biology ,Peripheral blood mononuclear cell ,Biochemistry ,Article ,Mice ,Mycosis Fungoides ,mir-223 ,Cell Line, Tumor ,hemic and lymphatic diseases ,Proto-Oncogenes ,microRNA ,medicine ,Animals ,Humans ,3' Untranslated Regions ,Molecular Biology ,Cell Proliferation ,Skin ,Mycosis fungoides ,Cell growth ,Gene Expression Profiling ,Cutaneous T-cell lymphoma ,Cell Biology ,medicine.disease ,Lymphoma, T-Cell, Cutaneous ,Lymphoma ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Disease Progression ,Leukocytes, Mononuclear ,NIH 3T3 Cells ,Cancer research - Abstract
The pathogenesis of the cutaneous T-cell lymphoma (CTCL), mycosis fungoides (MF), is unclear. MicroRNA (miRNA) are small noncoding RNAs that target mRNA leading to reduced mRNA translation. Recently, specific miRNA were shown to be altered in CTCL. We detected significantly reduced expression of miR-223 in early-stage MF skin, and further decreased levels of miR-223 in advanced-stage disease. CTCL peripheral blood mononuclear cells and cell lines also had reduced miR-223 as compared with controls. Elevated expression of miR-223 in these cell lines reduced cell growth and clonogenic potential, whereas inhibition of miR-223 increased cell numbers. Investigations into putative miR-223 targets with oncogenic function, including E2F1 and MEF2C , and the predicted miR-223 target, TOX , revealed that all three were targeted by miR-223 in CTCL. E2F1, MEF2C, and TOX proteins were decreased with miR-223 overexpression, whereas miR-223 inhibition led to increased protein levels in CTCL. In addition, we showed that the 3′-UTR of TOX mRNA was a genuine target of miR-223. Therefore, reduced levels of miR-223 in MF/CTCL lead to increased expression of E2F1, MEF2C, and TOX, which likely contributes to the development and/or progression of CTCL. Thus, miR-223 and its targets may be useful for the development of new therapeutics for MF/CTCL.
- Published
- 2014
- Full Text
- View/download PDF
27. Letter to the Editor regarding the paper by N. Azzam et al. ‘Germline polymorphisms on RET proto-oncogene involved in medullary thyroid carcinoma in a Druze family’
- Author
-
Klaus Brusgaard, Christian Godballe, and Jes Sloth Mathiesen
- Subjects
Cancer Research ,Proto-Oncogenes ,Letter ,Letter to the editor ,Medullary cavity ,business.industry ,Proto-Oncogene Proteins c-ret ,RET proto-oncogene ,Proto-Oncogene Mas ,Germline ,Thyroid carcinoma ,Oncology ,Cancer research ,Humans ,Medicine ,Thyroid Neoplasms ,business - Published
- 2018
28. DNMT3B7 expression related to MENT expression and its promoter methylation in human lymphomas
- Author
-
Lobna Alkebsi, Kunio Yanagisawa, Yoshihisa Nojima, Yoshiaki Ogawa, Yoshiko Sasaki, Takeki Mitsui, Takayuki Saitoh, Hiromi Koiso, Yohei Osaki, Hiroshi Handa, Norifumi Tsukamoto, Hikaru Hattori, Akihiko Yokohama, and Hirokazu Murakami
- Subjects
DNA (Cytosine-5-)-Methyltransferase 1 ,Cancer Research ,Lymphoma ,DNMT3B ,Biology ,Proto-Oncogene Mas ,DNA methyltransferase ,Gene Expression Regulation, Enzymologic ,DNA Methyltransferase 3A ,Proto-Oncogenes ,Promoter methylation ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Promoter Regions, Genetic ,Alternative splicing ,Hematology ,DNA Methylation ,Molecular biology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Isoenzymes ,Alternative Splicing ,Oncology ,embryonic structures ,Leukocytes, Mononuclear ,Function (biology) - Abstract
DNA methyltransferase (DNMT) 3B7 is the most expressed DNMT3B splice variant. It was reported that the loss of DNMT3B function led to overexpression of the MEthylated in Normal Thymocyes (MENT) and accelerated mouse lymphomagenesis. We investigated the DNMT3B7 expression and its relationship to MENT expression and promoter methylation in human lymphomas. DNMT3B7 and MENT expression were significantly (p0.0001, p0.01) higher in lymphomas than in non-malignant. Expression of DNMT3B7 and MENT were associated with MENT promoter hypomethylation. DNMT3B7 overexpression might interfere with the normal DNA methylation mechanism required for silencing the MENT proto-oncogene, and may accelerate human lymphomagenesis.
- Published
- 2013
29. SUMO1 negatively regulates the transcriptional activity of EVI1 and significantly increases its co-localization with EVI1 after treatment with arsenic trioxide
- Author
-
Anjan K. Pradhan, Sneha Singh, and Soumen Chakraborty
- Subjects
Transcription, Genetic ,Lysine ,SUMO protein ,Fluorescent Antibody Technique ,Apoptosis ,Proto-Oncogene Mas ,Antileukemic agent ,Arsenicals ,Immunoenzyme Techniques ,Transactivation ,chemistry.chemical_compound ,Arsenic Trioxide ,Arsenic trioxide (ATO) ,Tumor Cells, Cultured ,Arsenic trioxide ,Ovarian Neoplasms ,Reverse Transcriptase Polymerase Chain Reaction ,Oxides ,Cell biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Biochemistry ,Female ,Transcriptional Activation ,Acute promyelocytic leukemia ,Chromatin Immunoprecipitation ,Blotting, Western ,SUMO-1 Protein ,bcl-X Protein ,Antineoplastic Agents ,Biology ,Real-Time Polymerase Chain Reaction ,SIRT1 ,Proto-Oncogenes ,medicine ,Humans ,Immunoprecipitation ,RNA, Messenger ,Ligase activity ,Molecular Biology ,Bcl-xL ,Sumoylation ,Cell Biology ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,EVI1 ,PIASy ,chemistry ,Cancer cell ,Mutagenesis, Site-Directed ,Transcription Factors - Abstract
Aberrant expression of the proto-oncogene EVI1 (ecotropic virus integration site1) has been implicated not only in myeloid or lymphoid malignancies but also in colon, ovarian and breast cancers. Despite its importance in oncogenesis, the regulatory factors and mechanisms that potentiate the function of EVI1 and its consequences are partially known. Here we demonstrated that EVI1 is post-translationally modified by SUMO1 at lysine residues 533, 698 and 874. Although both EVI1 and SUMO1 were found to co-localize in nuclear speckles, the sumoylation mutant of EVI1 failed to co-localize with SUMO1. Sumoylation abrogated the DNA binding efficiency of EVI1 and also affected EVI1 mediated transactivation. The SUMO ligase PIASy was found to play a bi-directional role on EVI1, PIASy enhanced EVI1 sumoylation and augmented sumoylated EVI1 mediated repression. PIASy was also found to interact with EVI1 and impaired EVI1 transcriptional activity independent of its ligase activity. Arsenic trioxide (ATO) known to act as an antileukemic agent for acute promyelocytic leukemia (APL) not only enhanced EVI1 sumoylation but also enhanced the co-localization of EVI1 and SUMO1 in nuclear bodies distinct from PML nuclear bodies. ATO treatment also affected the Bcl-xL protein expression in EVI1 positive cell line. Thus, the results showed that arsenic treatment enhanced EVI1 sumoylation, deregulated Bcl-xL, which eventually may induce apoptosis in EVI1 positive cancer cells. The study for the first time explores and reports sumoylation of EVI1, which plays an essential role in regulating its function.
- Published
- 2013
30. EVI1 targets ΔNp63 and upregulates the cyclin dependent kinase inhibitor p21 independent of p53 to delay cell cycle progression and cell proliferation in colon cancer cells
- Author
-
Aditya Kumar Panda, Nivedita Kuila, Kasturi Bala Nayak, Alok Das Mohapatra, and Soumen Chakraborty
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Transcription, Genetic ,Molecular Sequence Data ,Down-Regulation ,Proto-Oncogene Mas ,Biochemistry ,Downregulation and upregulation ,Cyclin-dependent kinase ,Proto-Oncogenes ,medicine ,Humans ,Gene silencing ,Gene Silencing ,Promoter Regions, Genetic ,Cell Proliferation ,Base Sequence ,biology ,Cell growth ,Tumor Suppressor Proteins ,Cell Cycle ,Zinc Fingers ,Cell Biology ,Cell cycle ,HCT116 Cells ,medicine.disease ,Phenotype ,Molecular biology ,MDS1 and EVI1 Complex Locus Protein ,Protein Structure, Tertiary ,Up-Regulation ,Cell biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Leukemia ,HEK293 Cells ,Colonic Neoplasms ,biology.protein ,Tumor Suppressor Protein p53 ,HT29 Cells ,Function (biology) ,Protein Binding ,Transcription Factors - Abstract
Several lines of evidence suggest that specific transcriptional events are involved in cell cycle, proliferation and differentiation processes; however, their deregulation by proto-oncogenes are involved in the development of leukemia and tumors. One such proto-oncogene is ecotropic viral integration site I which can differentially effect cell cycle progression and proliferation, in cell types of different origin. Our data for the first time shows that ecotropic viral integration site I binds to ΔNp63 promoter element directly and down regulates its expression. Down regulation of ΔNp63 induces the expression of p21 in HT-29 cells and also in colon carcinoma cells that do not express p53 including patient samples expressing low level of p53, that eventually delay cell cycle progression at G0/G1 phase. Concomitant silencing of ecotropic viral integration site I from the cells or introduction of ΔNp63 to the cells significantly rescued this phenotype, indicating the growth defect induced by ΔNp63 deficiency to be, at least in part, attributable to ecotropic viral integration site I function. The mutual regulation between ecotropic viral integration site I and ΔNp63 may constitute a novel axis which might affect the downstream pathways in cells that do not express functional p53.
- Published
- 2013
31. Altered Ca2+ signaling in cancer cells: Proto-oncogenes and tumor suppressors targeting IP3 receptors
- Author
-
Haidar Akl and Geert Bultynck
- Subjects
Cancer Research ,Voltage-dependent anion channel ,bcl-X Protein ,Promyelocytic Leukemia Protein ,Mitochondrion ,Biology ,Neoplasms ,Proto-Oncogenes ,Genetics ,Animals ,Humans ,Inositol 1,4,5-Trisphosphate Receptors ,Calcium Signaling ,PI3K/AKT/mTOR pathway ,Tumor Suppressor Proteins ,Endoplasmic reticulum ,Microfilament Proteins ,Membrane Proteins ,Nuclear Proteins ,STIM1 ,Cell biology ,Proto-Oncogene Proteins c-bcl-2 ,Oncology ,Apoptosis ,Cancer cell ,biology.protein ,Myeloid Cell Leukemia Sequence 1 Protein ,Beclin-1 ,Apoptosis Regulatory Proteins ,Proto-Oncogene Proteins c-akt ,Homeostasis ,Transcription Factors - Abstract
Proto-oncogenes and tumor suppressors critically control cell-fate decisions like cell survival, adaptation and death. These processes are regulated by Ca 2 + signals arising from the endoplasmic reticulum, which at distinct sites is in close proximity to the mitochondria. These organelles are linked by different mechanisms, including Ca 2 + -transport mechanisms involving the inositol 1,4,5-trisphosphate receptor (IP 3 R) and the voltage-dependent anion channel (VDAC). The amount of Ca 2 + transfer from the endoplasmic reticulum to mitochondria determines the susceptibility of cells to apoptotic stimuli. Suppressing the transfer of Ca 2 + from the endoplasmic reticulum to the mitochondria increases the apoptotic resistance of cells and may decrease the cellular responsiveness to apoptotic signaling in response to cellular damage or alterations. This can result in the survival, growth and proliferation of cells with oncogenic features. Clearly, proper maintenance of endoplasmic reticulum Ca 2 + homeostasis and dynamics including its links with the mitochondrial network is essential to detect and eliminate altered cells with oncogenic features through the apoptotic pathway. Proto-oncogenes and tumor suppressors exploit the central role of Ca 2 + signaling by targeting the IP 3 R. There are an increasing number of reports showing that activation of proto-oncogenes or inactivation of tumor suppressors directly affects IP 3 R function and endoplasmic reticulum Ca 2 + homeostasis, thereby decreasing mitochondrial Ca 2 + uptake and mitochondrial outer membrane permeabilization. In this review, we provide an overview of the current knowledge on the proto-oncogenes and tumor suppressors identified as IP 3 R-regulatory proteins and how they affect endoplasmic reticulum Ca 2 + homeostasis and dynamics.
- Published
- 2013
32. Patterns of scAAV Vector Insertion Associated With Oncogenic Events in a Mouse Model for Genotoxicity
- Author
-
Kimberly Zaraspe, Douglas M. McCarty, Lucia E. Rosas, Krista M. D. La Perle, Jessica L. Grieves, and Haiyan Fu
- Subjects
Male ,Cell ,Mice, SCID ,medicine.disease_cause ,law.invention ,Mice ,Liver Neoplasms, Experimental ,0302 clinical medicine ,law ,Drug Discovery ,Vector (molecular biology) ,Mice, Inbred C3H ,0303 health sciences ,Dependovirus ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Recombinant DNA ,Molecular Medicine ,Female ,SOS1 Protein ,medicine.drug ,Transcriptional Activation ,Carcinoma, Hepatocellular ,DNA damage ,Virus Integration ,Fibroblast Growth Factor 3 ,Genetic Vectors ,Molecular Sequence Data ,Genome, Viral ,Biology ,Virus ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Proto-Oncogenes ,Genetics ,medicine ,Animals ,Hepatectomy ,Molecular Biology ,030304 developmental biology ,Pharmacology ,Severe combined immunodeficiency ,Base Sequence ,medicine.disease ,Mutagenesis, Insertional ,Commentary ,Cancer research ,Camptothecin ,Fibroblast Growth Factor 10 ,Genotoxicity ,DNA Damage - Abstract
Recombinant adeno-associated virus (rAAV) vectors have gained an extensive record of safety and efficacy in animal models of human disease. Infrequent reports of genotoxicity have been limited to specific vectors associated with excess hepatocellular carcinomas (HCC) in mice. In order to understand potential mechanisms of genotoxicity, and identify patterns of insertion that could promote tumor formation, we compared a self-complementary AAV (scAAV) vector designed to promote insertional activation (scAAV-CBA-null) to a conventional scAAV-CMV-GFP vector. HCC-prone C3H/HeJ mice and severe combined immunodeficiency (SCID) mice were infected with vector plus secondary treatments including partial hepatectomy (HPX) and camptothecin (CPT) to determine the effects of cell cycling and DNA damage on tumor incidence. Infection with either vector led to a significant increase in HCC incidence in male C3H/HeJ mice. Partial HPX after infection reduced HCC incidence in the cytomegalovirus-green fluorescent protein (CMV-GFP)-infected mice, but not in the cognate chicken β-actin (CBA)-null infected group. Tumors from CBA-null infected, hepatectomized mice were more likely to contain significant levels of vector DNA than tumors from the corresponding CMV-GFP-infected group. Most CBA-null vector insertions recovered from tumors were associated with known proto-oncogenes or tumor suppressors. Specific patterns of insertion suggested read-through transcription, enhancer effects, and disruption of tumor suppressors as likely mechanisms for genotoxicity.
- Published
- 2012
33. Prdm3 and Prdm16 are H3K9me1 Methyltransferases Required for Mammalian Heterochromatin Integrity
- Author
-
Danny Reinberg, Inês Pinheiro, Mineo Kurokawa, Nicholas Shukeir, Gerhard Mittler, Christoph Fritzsch, Christel Genoud, Raphaël Margueron, Susumu Goyama, Monika Lachner, Michael Eisold, Florian Richter, Jinsook Son, and Thomas Jenuwein
- Subjects
Methyltransferase ,Heterochromatin ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Gene Knockout Techniques ,Mice ,Proto-Oncogenes ,Gene expression ,Animals ,Humans ,Heterochromatin organization ,Genetics ,Nuclear Lamina ,Biochemistry, Genetics and Molecular Biology(all) ,EZH2 ,Histone-Lysine N-Methyltransferase ,Methylation ,Fibroblasts ,MDS1 and EVI1 Complex Locus Protein ,DNA-Binding Proteins ,Nuclear lamina ,Heterochromatin protein 1 ,HeLa Cells ,Transcription Factors - Abstract
SummaryHeterochromatin serves important functions, protecting genome integrity and stabilizing gene expression programs. Although the Suv39h methyltransferases (KMTs) are known to ensure pericentric H3K9me3 methylation, the mechanisms that initiate and maintain mammalian heterochromatin organization remain elusive. We developed a biochemical assay and used in vivo analyses in mouse embryonic fibroblasts to identify Prdm3 and Prdm16 as redundant H3K9me1-specific KMTs that direct cytoplasmic H3K9me1 methylation. The H3K9me1 is converted in the nucleus to H3K9me3 by the Suv39h enzymes to reinforce heterochromatin. Simultaneous depletion of Prdm3 and Prdm16 abrogates H3K9me1 methylation, prevents Suv39h-dependent H3K9me3 trimethylation, and derepresses major satellite transcription. Most strikingly, DNA-FISH and electron microscopy reveal that combined impairment of Prdm3 and Prdm16 results in disintegration of heterochromatic foci and disruption of the nuclear lamina. Our data identify Prdm3 and Prdm16 as H3K9me1 methyltransferases and expose a functional framework in which anchoring to the nuclear periphery helps maintain the integrity of mammalian heterochromatin.
- Published
- 2012
34. Intronic RET gene variants in Down syndrome–associated Hirschsprung disease in an African population
- Author
-
Monique G. Zaahl and Samuel W. Moore
- Subjects
Male ,Down syndrome ,Genotype ,DNA Mutational Analysis ,Population ,Black People ,Single-nucleotide polymorphism ,Comorbidity ,RET proto-oncogene ,Biology ,Polymorphism, Single Nucleotide ,White People ,South Africa ,Germline mutation ,Proto-Oncogenes ,medicine ,Humans ,Hirschsprung Disease ,education ,Total colonic aganglionosis ,Genetics ,education.field_of_study ,Proto-Oncogene Proteins c-ret ,Infant ,General Medicine ,medicine.disease ,Molecular biology ,Introns ,Causality ,Europe ,Intestines ,Pediatrics, Perinatology and Child Health ,Intronic SNP ,Female ,Surgery ,Down Syndrome ,Polymorphism, Restriction Fragment Length - Abstract
Background Clinical association between Hirschsprung disease (HD) and Down syndrome (DS) is well established. RET promoter and intron 1 variations have been shown to interfere with RET function, increasing the risk of HD pathogenesis. The intronic single-nucleotide polymorphism 2 (SNP2 [rs2435357]) has been associated with DS-associated HD (DS-HD). This study focuses on variations of specific RET intron, 1 SNPs (viz, SNP1 [rs2506004] and SNP2 [rs2435357]) in DS-HD. Patients and Methods DNA was extracted from paraffin-embedded tissue samples and whole blood in 14 patients with DS with histologically proven HD. Polymerase chain reaction products of RET intron 1 were screened for genetic variation and matched to DS and controls from the general population. Results Thirty-seven blood and/or tissue from 14 patients with DS-HD were investigated. RET intronic variations (SNP1 [rs2506004] or SNP2 [rs2435357]) were detected in all patients. SNP1 was detected in all patients, was heterozygous in 9, and homozygous in 5 samples (all aganglionic and 1 total colonic aganglionosis). SNP2 was absent in 6 patients, heterozygous in 6, and homozygous in 3. Three DS controls had a heterozygous SNP1. Homozygous intronic SNP RET variations were related to aganglionic tissue but not normally ganglionated or transitional zone from the same individual. Conclusions Potential disease-related RET mutations were identified in the intron region in 80% of patients with DS-HD investigated, suggesting a causal relationship. The presence of a homozygous form in the aganglionic tissue probably represents a somatic mutation, which suggests local microenvironmental factors in HD pathogenesis.
- Published
- 2012
35. Angiopoietin1 contributes to the maintenance of cell quiescence in EVI1high leukemia cells
- Author
-
Kazuko Kaneda, Yusuke Saito, Norio Yamakawa, Kazuhiro Morishita, and Emi Ichihara
- Subjects
Biophysics ,Biology ,Biochemistry ,Jurkat cells ,Mice ,Cell quiescence ,Cancer stem cell ,Cell Line, Tumor ,hemic and lymphatic diseases ,Proto-Oncogenes ,Angiopoietin-1 ,medicine ,Animals ,Cyclin-Dependent Kinase Inhibitor p18 ,Humans ,Molecular Biology ,ABL ,Gene Expression Regulation, Leukemic ,Cell Cycle ,Myeloid leukemia ,Cell Biology ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,DNA-Binding Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,Immunology ,Cancer research ,Drug Screening Assays, Antitumor ,Stem cell ,Transcription Factors - Abstract
Ecotropic viral integration site-1 (EVI1) is an oncogenic transcription factor in human acute myeloid leukemia (AML) associated with poor prognosis. Because the drug-resistance of leukemia cells is partly dependent on cell quiescence in the bone marrow niche, EVI1 may be involved in cell cycle regulation in leukemia cells. As a candidate regulator of the cell cycle in leukemia cells with high EVI1 expression (EVI1high), we analyzed angiopoietin1 (Ang1), which is a down-regulated gene in EVI1-deficient mice and is involved in the quiescence of hematopoietic stem cells. The results of real-time PCR analyses showed that Ang1 is highly expressed in leukemia cell lines and primary AML cells with EVI1high expression. Introduction of shRNA against EVI1 into EVI1high leukemia cells down-regulated Ang1 expression. Moreover, knockdown of Ang1 in EVI1high leukemia cells promoted cell cycle progression and down-regulated the CDK inhibitor p18 (INK4c). Treatment with a decoy Tie2/Fc protein also down-regulated the expression of p18. These results suggest that Ang1/Tie2 signaling may suppress cell cycle progression via maintenance of G0/G1 phase through up-regulation of p18 expression. This mechanism may help to maintain EVI1high leukemia cells in the bone marrow niche and promote resistance to anti-cancer drugs.
- Published
- 2011
36. The evolution of MDM2 family genes
- Author
-
Jamil Momand, Vladimir A. Belyi, and Alberto Villegas
- Subjects
Time Factors ,Protein domain ,Cell Cycle Proteins ,Biology ,Genome ,Article ,Conserved sequence ,Evolution, Molecular ,Gene Duplication ,Proto-Oncogene Proteins ,Proto-Oncogenes ,Gene duplication ,Gene cluster ,Genetics ,Animals ,Humans ,Gene family ,neoplasms ,Gene ,Conserved Sequence ,Concerted evolution ,Genetic Variation ,Nuclear Proteins ,Proto-Oncogene Proteins c-mdm2 ,General Medicine ,Genes, p53 ,Invertebrates ,Protein Structure, Tertiary ,Multigene Family ,Vertebrates - Abstract
MDM2 and MDM4 are proto-oncoproteins that bind to and inhibit members of the p53 protein family, p53, p73 and possibly p63. p53 is a mammalian tumor suppressor and p63 and p73 are critical for development. With the sequencing of genomes from multiple organisms there is mounting evidence for a consensus scenario of p53 gene family evolution. A single p53/p63/p73 gene is in invertebrates and required for maintenance of germline DNA. Gene duplication occurred in an ancestor in common with cartilaginous fishes, giving rise to a separate p53 gene and at least one ancestral p63/p73 gene. In bony vertebrates, all three p53 gene family paralogs, p53, p63, and p73 are distinct genes. This raises the question of how MDM2 and MDM4 genes evolved. We show evidence that MDM2 and MDM4 arose from a gene duplication event prior to the emergence of bony vertebrates more than 440 millionyears ago. Comparative genome studies indicate that invertebrate organisms have only one MDM homolog. In jawed vertebrates, the p53-binding domains of MDM2 and MDM4 proteins evolved at a high rate, approaching the evolution rate of the MDM2-binding domain of p53. However, the MDM2-binding domain of p73 exhibits markedly stronger conservation suggesting novel p53-independent functions. The most conserved domain within all MDM2 family members is the RING domain of the MDM2 ortholog which is responsible for ubiquitination of p53 and heterodimerization with MDM4. We suggest a model where oligomerization is an ancient function of MDM and ubiquitination activity was acquired later near the MDM gene duplication event coinciding with the time of the emergence of p53 as a distinct gene.
- Published
- 2011
37. Discovery of novel imidazo[1,2-a]pyrazin-8-amines as Brk/PTK6 inhibitors
- Author
-
Yan Wang, Patrick J. Curran, Jack B. Bracken, Gerald W. Shipps, M. Arshad Siddiqui, Hua Miao, David B. Belanger, Hongbo Zeng, and Michael Malkowski
- Subjects
Clinical Biochemistry ,High selectivity ,Dasatinib ,Pharmaceutical Science ,Breast Neoplasms ,Protein Serine-Threonine Kinases ,Biochemistry ,Inhibitory Concentration 50 ,Mice ,Structure-Activity Relationship ,Aurora Kinases ,Proto-Oncogenes ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Computer Simulation ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Dose-Response Relationship, Drug ,Chemistry ,Kinase ,Drug discovery ,Organic Chemistry ,Imidazoles ,RNA-Binding Proteins ,Signal transducing adaptor protein ,Oncogenes ,Protein-Tyrosine Kinases ,Neoplasm Proteins ,DNA-Binding Proteins ,Thiazoles ,Phenotype ,Pyrimidines ,Drug Design ,Pyrazines ,Melanocytes ,Molecular Medicine ,Female ,PTK6 ,Drug Screening Assays, Antitumor ,Tyrosine kinase ,medicine.drug - Abstract
A series of substituted imidazo[1,2-a]pyrazin-8-amines were discovered as novel breast tumor kinase (Brk)/protein tyrosine kinase 6 (PTK6) inhibitors. Tool compounds with low-nanomolar Brk inhibition activity, high selectivity towards other kinases and desirable DMPK properties were achieved to enable the exploration of Brk as an oncology target.
- Published
- 2011
38. Apoptosis in polycystic kidney disease
- Author
-
Beatrice Goilav
- Subjects
Programmed cell death ,TRPP Cation Channels ,medicine.medical_treatment ,Apoptosis ,Disease ,Mice ,Polycystic kidney disease ,Proto-Oncogenes ,medicine ,Animals ,Humans ,Molecular Biology ,Caspase ,Polycystic Kidney Diseases ,Models, Statistical ,biology ,TOR Serine-Threonine Kinases ,Cell Cycle ,Cell cycle ,medicine.disease ,Caspase Inhibitors ,Cyclin-Dependent Kinases ,Cytokine ,Caspases ,Immunology ,Cancer research ,biology.protein ,Molecular Medicine ,Signal Transduction ,Kidney disease - Abstract
Apoptosis is the process of programmed cell death. It is a ubiquitous, controlled process consuming cellular energy and designed to avoid cytokine release despite activation of local immune cells, which clear the cell fragments. The process occurs during organ development and in maintenance of homeostasis. Abnormalities in any step of the apoptotic process are associated with autoimmune diseases and malignancies. Polycystic kidney disease (PKD) is the most common inherited kidney disease leading to end-stage renal disease (ESRD). Cyst formation requires multiple mechanisms and apoptosis is considered one of them. Abnormalities in apoptotic processes have been described in various murine and rodent models of PKD as well as in human PKD kidneys. The purpose of this review is to outline the role of apoptosis in progression of PKD as well as to describe the mechanisms involved. This article is part of a Special Issue entitled: Polycystic Kidney Disease.
- Published
- 2011
39. Frequent EVI1 translocations in myeloid blast crisis CML that evolves through tyrosine kinase inhibitors
- Author
-
Meenal Chalukya, Charles L. Sawyers, P. Nagesh Rao, John Nicoll, Neil P. Shah, Elizabeth Spiteri, Ronald Paquette, David Elashoff, and Gouri Nanjangud
- Subjects
Adult ,Male ,Cancer Research ,Myeloid ,DNA Repair ,medicine.drug_class ,Blotting, Western ,Fusion Proteins, bcr-abl ,Chromosomal translocation ,Genes, abl ,Biology ,Translocation, Genetic ,Tyrosine-kinase inhibitor ,Evolution, Molecular ,Myelogenous ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Proto-Oncogenes ,Genetics ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Protein Kinase Inhibitors ,Molecular Biology ,In Situ Hybridization, Fluorescence ,Aged ,ABL ,medicine.diagnostic_test ,Myeloid leukemia ,Middle Aged ,Protein-Tyrosine Kinases ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,respiratory tract diseases ,DNA-Binding Proteins ,Leukemia ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Mutation ,Immunology ,Cancer research ,Female ,Blast Crisis ,Transcription Factors ,Fluorescence in situ hybridization - Abstract
Clinical variables associated with ecotropic viral integration site 1 (EVI1) translocations were evaluated in 42 consecutive chronic myeloid leukemia (CML) patients in myeloid blast crisis (MBC). Translocations were confirmed with fluorescence in situ hybridization, and Western blot analysis demonstrated EVI1 expression. Translocations of EVI1 were present in 3 of 24 (12%) patients whose disease evolved MBC before tyrosine kinase inhibitor (TKI) exposure, and 7 of 18 (39%) patients who had received one or more TKIs. Univariate analysis showed that prior TKI therapy was the only clinical variable that was significantly associated with EVI1 translocation (P = 0.047). TKI-resistant BCR-ABL1 mutations were present in 71% of MBC patients with EVI1 translocations at the time of disease progression. These observations suggest that EVI1 overexpression collaborates with BCR-ABL1 in the evolution of TKI-resistant MBC. Inhibition of c-ABL kinase-mediated DNA double-strand repair by TKIs may predispose to EVI1 translocation in this setting.
- Published
- 2011
40. EVI1 up-regulates the stress responsive gene SIRT1 which triggers deacetylation and degradation of EVI1
- Author
-
Soumen Chakraborty, Anjan K. Pradhan, Sneha Singh, and Nivedita Kuila
- Subjects
Transcriptional Activation ,Chromatin Immunoprecipitation ,Blotting, Western ,Biophysics ,Biology ,Biochemistry ,Mice ,Sirtuin 1 ,Structural Biology ,Cell Line, Tumor ,Proto-Oncogenes ,Genetics ,Animals ,Humans ,Luciferase ,Promoter Regions, Genetic ,Molecular Biology ,Zinc finger ,Reverse Transcriptase Polymerase Chain Reaction ,Myeloid leukemia ,Acetylation ,Promoter ,Molecular biology ,MDS1 and EVI1 Complex Locus Protein ,Up-Regulation ,DNA-Binding Proteins ,HEK293 Cells ,PCAF ,RNA Interference ,Histone deacetylase ,Tumor Suppressor Protein p53 ,K562 Cells ,Chromatin immunoprecipitation ,Protein Binding ,Transcription Factors - Abstract
EVI1 (Ecotropic Viral Integration site I), which was originally identified as a site of viral integration in murine myeloid tumors, encodes a complex protein required for embryogenesis. The gene is known to express inappropriately in many types of human myeloid leukemias and solid tumors. Forced expression of EVI1 in murine hematopoietic precursor cells lead to abnormal differentiation and increased proliferation. EVI1 encodes two sets of zinc finger domains due to which it behaves as a transcriptional factor. However, except a few, the targets of EVI1 are not well understood and hence also the mechanism by which it initiates oncogenesis is not very clear. In this report, we show that SIRT1, a histone deacetylase is a direct target of EVI1. In vivo chromatin immunoprecipitation assay revealed that EVI1 binds to the promoter region of SIRT1 approximately 1 kb upstream of the transcription start site. The functionality of the site was deduced by luciferase assay which showed that EVI1 significantly increases the SIRT1 promoter activity. SIRT1 was also found to be up regulated in cell lines and in chronic myeloid leukemia patient samples where EVI1 was detected. Over expression of SIRT1 in cells shows that it interacts with EVI1 and this interaction lead to the deacetylation of the protein. Upon deacetylation the stability of EVI1 was found to be affected which was negatively regulated by nicotinamide (NAM). Our results thus identify an EVI1-SIRT1 axis in the regulation of EVI1 activity suggesting a possible role of SIRT1 in EVI1 positive neoplasms.
- Published
- 2011
41. SnoN in mammalian development, function and diseases
- Author
-
Kunxin Luo and Nadine Jahchan
- Subjects
Embryonic Development ,Biology ,medicine.disease_cause ,Proto-Oncogene Mas ,Article ,Mice ,Transforming Growth Factor beta ,Proto-Oncogene Proteins ,Proto-Oncogenes ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Pharmacology ,Regulation of gene expression ,Activator (genetics) ,Cell growth ,Intracellular Signaling Peptides and Proteins ,Transforming growth factor beta ,Genes, p53 ,Embryonic stem cell ,Cell biology ,Cell Transformation, Neoplastic ,Gene Expression Regulation ,Trans-Activators ,biology.protein ,Female ,Signal transduction ,Carcinogenesis ,Signal Transduction ,Transforming growth factor - Abstract
SnoN (Ski-novel protein) was discovered as a nuclear proto-oncogene on the basis of its ability to induce transformation of chicken and quail embryonic fibroblasts. As a crucial negative regulator of transforming growth factor-β (TGF-β) signaling and also an activator of p53, it plays an important role in regulating cell proliferation, senescence, apoptosis, and differentiation. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed important functions of SnoN in normal epithelial development and tumorigenesis. Evidence suggests that SnoN has both pro-oncogenic and anti-oncogenic functions by modulating multiple signaling pathways. These studies suggest that SnoN may have broad functions in the development and homeostasis of embryonic and postnatal tissues.
- Published
- 2010
42. The Proto-oncogene c-Kit Inhibits Tumor Growth by Behaving as a Dependence Receptor
- Author
-
Yan Sun, Andrea Paradisi, David Goldschneider, Patrick Mehlen, Laetitia Mazelin, Servane Tauszig-Delamasure, Amina Boussouar, Hong Wang, State Key Laboratory of Loess & Quaternary Geology, Laboratoire de Biologie Moléculaire de la Cellule (LBMC), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Institut de biologie moléculaire et cellulaire (IBMC), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), Développement Cancer et Thérapies Ciblées [Lyon] (LabEx DEVweCAN), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon, and Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Apoptosis ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Stem cell factor ,Mice, SCID ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Dependence receptor ,Proto-Oncogene Mas ,Tropomyosin receptor kinase C ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Proto-Oncogenes ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Phosphorylation ,Kinase activity ,Receptor ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,Stem Cell Factor ,biology ,Kinase ,Cell Biology ,3. Good health ,Proto-Oncogene Proteins c-kit ,030104 developmental biology ,Tumor progression ,biology.protein ,Cancer research ,Female ,Cell Division - Abstract
C-Kit has been generally considered as a receptor tyrosine kinase and a proto-oncogene, whose upregulation and mutation lead to tumor progression through its kinase activity. Clinically, drugs targeting the kinase activity of c-Kit, such as Imatinib (Gleevec), have been wildly used to treat patients with c-Kit related diseases. While the role of c-Kit as a proto-oncogene is of no doubt, some research reports and database analysis do not fit well the tumor promoting role of c-Kit, indicating a possible different role of c-Kit in cancer. Here, we show that c-Kit belongs to the dependence receptor family, similarly to other receptor tyrosine kinases such as MET, RET and TrkC. In the absent of its ligand SCF (stem cell factor), instead of staying inactive, c-Kit triggers apoptosis, which can be enhanced by silencing its kinase activity. Besides, we have shown that c-Kit is able to bind and activate caspase-9. Moreover, similarly to other dependence receptors, c-Kit is also cleaved by caspases-like protease at aspartic acid residue D816, which is crucial for its pro-apoptotic activity. The mutation of D816 site inhibits the c-Kit/caspase-9 binding and silences the pro-apoptotic activity of c-Kit. Of interest, c-Kit D816 mutation is one of the most common mutation of this receptor in many c-Kit related cancers and it promotes resistance against Gleevec treatment. We also show that overexpression of kinase mutated c-Kit is able to inhibit tumor growth in animal models, while the mutation of D816 site impairs the tumor suppressing activity. Furthermore, we develop a tool to block the SCF/c-Kit interaction, which unleashes the pro-apoptotic activity of c-Kit in cancers expressing this receptor. By using the pro-apoptotic activity of c-Kit, in combination with kinase inhibitors like Gleevec, we propose a novel therapeutic strategy. In conclusion, we demonstrate that c-Kit is a member of dependence receptor family, harboring intrinsic pro-apoptotic activity, which can be used as an alternative tool in cancer treatment
- Published
- 2018
43. Acetylation of Lysine 564 Adjacent to the C-terminal Binding Protein-binding Motif in EVI1 Is Crucial for Transcriptional Activation of GATA2
- Author
-
Akiko Shimahara, Ichiro Nishikata, Norio Yamakawa, and Kazuhiro Morishita
- Subjects
Transcriptional Activation ,Transcription, Genetic ,Amino Acid Motifs ,Biology ,Biochemistry ,DNA-binding protein ,Transcription (biology) ,Cell Line, Tumor ,Chlorocebus aethiops ,Proto-Oncogenes ,Animals ,Humans ,Gene Regulation ,Molecular Biology ,Transcription factor ,Regulation of gene expression ,Histone Acetyltransferase p300 ,Leukemia ,Lysine ,Binding protein ,GATA2 ,Zinc Fingers ,Promoter ,Cell Biology ,Molecular biology ,MDS1 and EVI1 Complex Locus Protein ,Protein Structure, Tertiary ,DNA-Binding Proteins ,GATA2 Transcription Factor ,Gene Expression Regulation, Neoplastic ,COS Cells ,Transcription Factors - Abstract
Ecotropic viral integration site 1 (EVI1) is an important transcription factor for leukemogenesis. EVI1 is a member of a group of transcription factors with C-terminal binding protein (CtBP)-binding motifs that act as transcriptional co-repressors; however, we recently found that EVI1 directly activates GATA2 transcription, which is an important gene for the maintenance of hematopoietic stem cells. We show here that EVI1-activated GATA2 transcripts derive from exon 1S of GATA2, which is specifically activated in neural and hematopoietic cells. EVI1 was acetylated by the histone acetyltransferase p300/CBP association factor (P/CAF) in myeloid leukemia cells and hematopoietic progenitor cells. Acetylation at Lys(564), which is adjacent to the CtBP-binding consensus sequence of EVI1, was found to be important for transcriptional activation of GATA2. Mutation of Lys(564) to alanine (K564A) markedly reduced the ability of EVI1 to bind DNA and activate transcription of GATA2. Furthermore, we confirmed that Lys(564) in EVI1 was specifically acetylated in leukemia and primary hematopoietic cells by using an antibody directed against an acetylated Lys(564) EVI1 peptide. Moreover, co-transfection of P/CAF with EVI1 overcame the suppressive effect of the CtBP co-repressor and resulted in GATA2 transcriptional activation; nonetheless, CtBP2 was still included in the protein complex with EVI1 and P/CAF on the EVI1-binding site in the GATA2 promoter region. Thus, acetylation of EVI1 at Lys(564) by P/CAF enhances the DNA binding capacity of EVI1 and thereby contributes to the activation of GATA2.
- Published
- 2010
44. Systematic Characterization of Stress-Induced RNA Granulation
- Author
-
Yu Mi Woo, Sim Namkoong, Hojoong Kwak, Allison Ho, and Jun Hee Lee
- Subjects
0301 basic medicine ,Arsenites ,Biology ,Cytoplasmic Granules ,Article ,Transcriptome ,Mice ,03 medical and health sciences ,Stress granule ,Proto-Oncogenes ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,Ribonucleoprotein ,AU Rich Elements ,AU-rich element ,Messenger RNA ,Binding Sites ,Endoplasmic reticulum ,RNA-Binding Proteins ,RNA ,Cell Biology ,Endoplasmic Reticulum Stress ,HCT116 Cells ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Ribonucleoproteins ,Solubility ,NIH 3T3 Cells ,Unfolded protein response ,Thapsigargin ,RNA, Long Noncoding ,Heat-Shock Response ,Protein Binding ,Transcription Factors - Abstract
Summary Upon stress, cytoplasmic mRNA is sequestered to insoluble ribonucleoprotein (RNP) granules, such as the stress granule (SG). Partially due to the belief that translationally suppressed mRNAs are recruited to SGs in bulk, stress-induced dynamic redistribution of mRNA has not been thoroughly characterized. Here, we report that endoplasmic reticulum (ER) stress targets only a small subset of translationally suppressed mRNAs into the insoluble RNP granule fraction (RG). This subset, characterized by extended length and adenylate-uridylate (AU)-rich motifs, is highly enriched with genes critical for cell survival and proliferation. This pattern of RG targeting was conserved for two other stress types, heat shock and arsenite toxicity, which induce distinct responses in the total cytoplasmic transcriptome. Nevertheless, stress-specific RG-targeting motifs, such as guanylate-cytidylate (GC)-rich motifs in heat shock, were also identified. Previously underappreciated, transcriptome profiling in the RG may contribute to understanding human diseases associated with RNP dysfunction, such as cancer and neurodegeneration.
- Published
- 2018
45. Rosuvastatin inhibits norepinephrine-induced cardiac hypertrophy via suppression of Gh
- Author
-
Min Ji Cha, Namsik Chung, Eui-Young Choi, Byeong-Wook Song, Hye Jung Kim, Yangsoo Jang, Soyeon Lim, Eun-Ju Choi, Ki-Chul Hwang, and Woochul Chang
- Subjects
medicine.medical_specialty ,Myosin Light Chains ,G protein ,Intracellular Space ,Down-Regulation ,Cardiomegaly ,Transfection ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Muscle hypertrophy ,Rats, Sprague-Dawley ,Norepinephrine ,GTP-binding protein regulators ,GTP-Binding Proteins ,Internal medicine ,Proto-Oncogenes ,medicine ,Animals ,Myocytes, Cardiac ,Rosuvastatin ,RNA, Messenger ,RNA, Small Interfering ,Rosuvastatin Calcium ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase A ,Protein kinase C ,Pharmacology ,Sulfonamides ,Base Sequence ,biology ,Cell Membrane ,nutritional and metabolic diseases ,Rats ,Fluorobenzenes ,Protein Transport ,Pyrimidines ,Endocrinology ,HMG-CoA reductase ,biology.protein ,Calcium ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Cardiac Myosins ,medicine.drug - Abstract
Statins have recently been shown to produce anti-cardiac hypertrophic effects via the regulation of small GTPases. However, the effects of statins on G protein-mediated cardiac hypertrophy, which is the main pathway of cardiac hypertrophy, have not yet been studied. We sought to evaluate whether statin treatment directly suppresses cardiac hypertrophy through a large G protein-coupled pathway regardless of the regulation of small GTPases. Using neonatal rat cardiomyocytes, we evaluated norepinephrine-induced cardiac hypertrophy for suppressibility of rosuvastatin and the pathways involved by analyzing total protein/DNA content, cell surface area, immunoblotting and RT-PCR for the signal transduction molecule. In a concentration-dependent manner, rosuvastatin inhibited total protein synthesis and downregulated basal and norepinephrine-induced expressions of myosin light chain2 and the c-fos proto-oncogene in cardiomyocytes. Treatment with norepinephrine induced cardiac hypertrophy accompanied by G(h) expression and membrane translocation. Rosuvastatin inhibited G(h) protein activity in cardiomyocytes by inhibiting basal and norepinephrine-stimulated mRNA transcription, protein expression and membrane translocation; however, norepinephrine-stimulated G(q) protein expression was not inhibited. In addition, the norepinephrine-stimulated protein kinase C (PKC)-mitogen-activated protein kinase (MEK 1,2)-extracellular signal-regulated kinases (ERKs) signaling cascade was inhibited by pretreatment with rosuvastatin. Rosuvastatin treatment also helped maintain expression levels of SERCA2a and intracellular calcium concentration. G(h) protein is a novel target of statins in myocardial hypertrophy, and statin treatment may directly suppress cardiac hypertrophy through a large G(h) protein-coupled pathway regardless of the regulation of small GTPases.
- Published
- 2010
46. Insertional Transformation of Hematopoietic Cells by Self-inactivating Lentiviral and Gammaretroviral Vectors
- Author
-
Martijn H. Brugman, Axel Schambach, Christopher Baum, Daniela Zychlinski, Anne Galy, Juan A. Bueren, Tobias Maetzig, Ute Modlich, Sabine Charrier, Adrian J. Thrasher, Susana Navarro, and S Knoess
- Subjects
Mutant ,Genetic Vectors ,Bone Marrow Cells ,Biology ,Polymerase Chain Reaction ,03 medical and health sciences ,Transduction (genetics) ,Mice ,0302 clinical medicine ,Transduction, Genetic ,Proto-Oncogenes ,Drug Discovery ,Genetics ,Animals ,Gene ,Molecular Biology ,030304 developmental biology ,Regulator gene ,Regulation of gene expression ,Pharmacology ,0303 health sciences ,Lentivirus ,Intron ,Original Articles ,biology.organism_classification ,Blotting, Northern ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Mutagenesis, Insertional ,030220 oncology & carcinogenesis ,Molecular Medicine ,Gammaretrovirus - Abstract
Gene transfer vectors may cause clonal imbalance and even malignant cell transformation by insertional upregulation of proto-oncogenes. Lentiviral vectors (LV) with their preferred integration in transcribed genes are considered less genotoxic than gammaretroviral vectors (GV) with their preference for integration next to transcriptional start sites and regulatory gene regions. Using a sensitive cell culture assay and a series of self-inactivating (SIN) vectors, we found that the lentiviral insertion pattern was approximately threefold less likely than the gammaretroviral to trigger transformation of primary hematopoietic cells. However, lentivirally induced mutants also showed robust replating, in line with the selection for common insertion sites (CIS) in the first intron of the Evi1 proto-oncogene. This potent proto-oncogene thus represents a CIS for both GV and LV, despite major differences in their integration mechanisms. Altering the vectors' enhancer–promoter elements had a greater effect on safety than the retroviral insertion pattern. Clinical grade LV expressing the Wiskott–Aldrich syndrome (WAS) protein under control of its own promoter had no transforming potential. Mechanistic studies support the conclusion that enhancer-mediated gene activation is the major cause for insertional transformation of hematopoietic cells, opening rational strategies for risk prevention.
- Published
- 2009
- Full Text
- View/download PDF
47. Regulation by c-Myc of ncRNA expression
- Author
-
Robert J. White and Niall S. Kenneth
- Subjects
Regulation of gene expression ,Genetics ,Proto-Oncogenes ,RNA, Untranslated ,Genes, myc ,RNA ,Biology ,Non-coding RNA ,Models, Biological ,Proto-Oncogene Mas ,Proto-Oncogene Proteins c-myc ,Gene Expression Regulation ,microRNA ,Transfer RNA ,Animals ,Humans ,Gene ,Transcription factor ,Developmental Biology - Abstract
Deregulated activity of the proto-oncogene product c-Myc is instrumental in promoting many human cancers. As it is a transcription factor, priority has been given to identifying the genes that it regulates. Until recently, all the attention was focused on protein-encoding genes. It is now clear, however, that c-Myc also controls the production of many non-coding (nc) RNAs, including tRNA, rRNA and miRNAs. This involves it regulating the transcriptional activity of three different RNA polymerases. These ncRNAs are likely to contribute substantially to the complex biology and pathology that is associated with c-Myc.
- Published
- 2009
48. Acute promyelocytic leukemia relapsing as secondary acute myelogenous leukemia with translocation t(3;21)(q26;q22) and RUNX1–MDS1–EVI1 fusion transcript
- Author
-
Jaewoo Song, Jong Rak Choi, Yoo Hong Min, Juwon Kim, Sue Jung Kim, Sul Hee Yoon, Ohgun Kwon, and Tae Sung Park
- Subjects
Acute promyelocytic leukemia ,Cancer Research ,Myeloid ,Oncogene Proteins, Fusion ,Chromosomes, Human, Pair 21 ,Tretinoin ,Biology ,Translocation, Genetic ,Chromosome Painting ,Myelogenous ,chemistry.chemical_compound ,Promyelocytic leukemia protein ,Leukemia, Promyelocytic, Acute ,Bone Marrow ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Proto-Oncogenes ,Genetics ,medicine ,Humans ,neoplasms ,Molecular Biology ,Acute leukemia ,Neoplasms, Second Primary ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,MDS1 and EVI1 Complex Locus Protein ,Neoplasm Proteins ,DNA-Binding Proteins ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,RUNX1 ,chemistry ,Fusion transcript ,Karyotyping ,Core Binding Factor Alpha 2 Subunit ,biology.protein ,Cancer research ,Female ,Chromosomes, Human, Pair 3 ,Transcription Factors - Abstract
Acute promyelocytic leukemia (APL) is a subtype of acute myelogenous leukemia (AML) that is characterized by peculiar clinical and biologic features, including severe hemorrhagic diathesis, specific recurrent chromosomal aberration, and distinct morphologic features with predominant pathologic promyelocytes. A reciprocal translocation involving chromosomes 15 and 17, t(15;17)(q22;q21), is a characteristic feature of APL that represents approximately 5-8% of AML. The rearranged gene created by this translocation encodes a chimeric protein PML-RARA that is a transcriptional repressor. In contrast to other AML subtypes, APL is particularly sensitive to treatment with all trans-retinoic acid (ATRA) combined with chemotherapy, converting this once fatal leukemia to a highly curable disease. Nonetheless, therapy-related myelodysplastic syndrome-acute myelogenous leukemia (t-MDS/AML) has been reported as a rare complication of chemotherapy in APL. Of 30 APL cases described as t-MDS/AML in the literature, only 1 case relapsed as acute leukemia with t(3;21)(q26;q22). Here we describe a rare case of APL relapsing as secondary AML with t(3;21)(q26;q22) and clinically characterize this patient using the RUNX1 (previously AML1)-MDS1-EVI1 fusion transcript (with follow-up for 55 months), and review the relevant literature.
- Published
- 2008
49. Evi-1 Is a Critical Regulator for Hematopoietic Stem Cells and Transformed Leukemic Cells
- Author
-
Go Yamamoto, Seishi Ogawa, Mineo Kurokawa, Munetake Shimabe, Shigeru Chiba, Tomohiko Sato, Susumu Goyama, and Motoshi Ichikawa
- Subjects
Cell Survival ,medicine.medical_treatment ,Gene Dosage ,Bone Marrow Cells ,Hematopoietic stem cell transplantation ,Biology ,Mice ,Proto-Oncogenes ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,Cell Line, Transformed ,Cell Proliferation ,Mice, Knockout ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematopoietic Stem Cells ,Microarray Analysis ,medicine.disease ,Receptor, TIE-2 ,STEMCELL ,Embryonic stem cell ,MDS1 and EVI1 Complex Locus Protein ,Hematopoiesis ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Leukemia, Myeloid ,Immunology ,Molecular Medicine ,Bone marrow ,Stem cell ,Transcription Factors ,Adult stem cell - Abstract
SummaryEvi-1 has been recognized as one of the dominant oncogenes associated with murine and human myeloid leukemia. Here, we show that hematopoietic stem cells (HSCs) in Evi-1-deficient embryos are severely reduced in number with defective proliferative and repopulating capacity. Selective ablation of Evi-1 in Tie2+ cells mimics Evi-1 deficiency, suggesting that Evi-1 function is required in Tie2+ hematopoietic stem/progenitors. Conditional deletion of Evi-1 in the adult hematopoietic system revealed that Evi-1-deficient bone marrow HSCs cannot maintain hematopoiesis and lose their repopulating ability. In contrast, Evi-1 is dispensable for blood cell lineage commitment. Evi-1+/− mice exhibit the intermediate phenotype for HSC activity, suggesting a gene dosage requirement for Evi-1. We further demonstrate that disruption of Evi-1 in transformed leukemic cells leads to significant loss of their proliferative activity both in vitro and in vivo. Thus, Evi-1 is a common and critical regulator essential for proliferation of embryonic/adult HSCs and transformed leukemic cells.
- Published
- 2008
50. The gene structure of the Drosophila melanogaster proto-oncogene, kayak, and its nested gene, fos-intronic gene
- Author
-
Elliott S. Goldstein and Stephanie Gidget Hudson
- Subjects
Gene isoform ,Leucine zipper ,Codon, Initiator ,Biology ,Proto-Oncogene Mas ,Transcription (biology) ,Proto-Oncogenes ,Genetics ,Animals ,Drosophila Proteins ,Protein Isoforms ,Cloning, Molecular ,Gene ,Reverse Transcriptase Polymerase Chain Reaction ,Promoter ,General Medicine ,DNA-binding domain ,biology.organism_classification ,Molecular biology ,Introns ,Nested gene ,Drosophila melanogaster ,Mutation ,5' Untranslated Regions - Abstract
We present herein a new model for the structure of the Drosophila kayak gene as well as preliminary data on the functional differences of its various isoforms. kayak is a homolog of the human proto-oncogene, c-fos . kayak has three different starts of transcription, and therefore promoters (P) kay-α , (P) kay-β and (P) kay-γ . These three promoters lead to four different transcripts: kay-α , kay sro , kay-β and kay-γ . (P) kay-α produces two different transcripts: kay-α and kay sro where the other two promoters, (P) kay-β and (P) kay-γ , produce a single transcript each. The transcripts kay-α , β and γ all splice into the mainbody of the kay gene, which codes for the DNA binding domain and leucine zipper; kay sro is not spliced. Also, within this region is a nested gene, fos-intronic gene (fig) which is transcribed in the opposite direction. fig codes for a predicted PP2C phosphatase. fig has two different promoters which produce two different transcripts, both in the same reading frame, fig-α and β . This is an unusual gene structure for Drosophila . Only 13% of Drosophila genes have multiple promoters and only 7% have a nested gene. RT-PCR was performed on each transcript to determine the relative amounts of each RNA produced. All spliced kay transcripts appear to have equal abundance. The unspliced kay sro transcript has a lower abundance than kay-α . Both fig transcripts are also detected in all stages tested. Lethal phase analysis and complementation testing suggest that the three isoforms of kayak may have different functions.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.