18 research outputs found

    Silencing of HN1L suppresses the proliferation and migration of cancer cells

    Get PDF
    Background and purpose: HN1L is a member of the HN1 gene family and shares about 30% similarity with HN1 which is another member of the family on the primary protein sequence. Since HN1 is an important gene that is involved in various cellular mechanisms and also differentially expressed in carcinogenesis, we investigated the effect of HN1L on some malignant behaviors of various cancer cells.Material and methods: Co-expression analysis, Gene Ontology enrichment, and database searches were performed to predict the cellular roles of HN1, and to investigate its expression in cancers and their corresponding normal tissues. Western blotting and Real-Time PCR were used to compare the expression of HN1L in the normal prostate cells and prostate cancer cells. Cell proliferation and migration assays were used to investigate the effects of HN1L depletion on cell proliferation and migration.Results: The results of co-expression and Gene Ontology enrichment analyses showed that HN1L is co-expressed with DNA replication and DNA damage response/repair associated genes. The database search results revealed that HN1L expression increases in at least 10 diverse cancer types compared to their normal corresponding tissues. This result was confirmed in the prostate cancer cell model, experimentally. Silencing of HN1L inhibited proliferative and migrative behaviors of prostate, breast, colon, and cervix cancer cells.Conclusions: HN1L probably is a novel proto-oncogene that is involved in the DNA metabolism-related mechanisms, and high HN1L level promotes further proliferation and migration in the cancer cells

    Meta-analysis of the cell cycle related C12orf48

    Get PDF
    ABSTRACT: The cell cycle is a conserved process from yeast to mammals and focuses on mechanisms that regulate the timing and frequency of DNA replication and cell division. The temporal and spatial expression of the genes is tightly regulated to ensure accurate replication and transmission of DNA to daughter cells during the cycle. Although the genes involved in interphase are well studied, most of the genes which are involved in mitotic events still remain unidentified. Since, the discovery of mitosis related genes is still incomplete, we performed a co-expression and gene ontology analysis for revealing novel mitosis regulated genes. In this study, we showed that C12orf48 is co-expressed with well-known mitotic genes. Moreover, it is also co-expressed with the genes that have roles in interphase such as DNA replication. Furthermore, our results showed that C12orf48 is also differentially expressed in various cancers. Therefore, the results presented in this study suggest that C12orf48 may be an important molecule for both interphase and mitosis. Since, the molecules involved in these mechanisms are crucial for proliferation as well as in carcinogenesis, C12orf48 should be considered as a novel cell cycle and carcinogenesis related gene

    European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS).

    Get PDF
    The European Cooperation in Science and Technology (COST) provides an ideal framework to establish multi-disciplinary research networks. COST Action BM1203 (EU-ROS) represents a consortium of researchers from different disciplines who are dedicated to providing new insights and tools for better understanding redox biology and medicine and, in the long run, to finding new therapeutic strategies to target dysregulated redox processes in various diseases. This report highlights the major achievements of EU-ROS as well as research updates and new perspectives arising from its members. The EU-ROS consortium comprised more than 140 active members who worked together for four years on the topics briefly described below. The formation of reactive oxygen and nitrogen species (RONS) is an established hallmark of our aerobic environment and metabolism but RONS also act as messengers via redox regulation of essential cellular processes. The fact that many diseases have been found to be associated with oxidative stress established the theory of oxidative stress as a trigger of diseases that can be corrected by antioxidant therapy. However, while experimental studies support this thesis, clinical studies still generate controversial results, due to complex pathophysiology of oxidative stress in humans. For future improvement of antioxidant therapy and better understanding of redox-associated disease progression detailed knowledge on the sources and targets of RONS formation and discrimination of their detrimental or beneficial roles is required. In order to advance this important area of biology and medicine, highly synergistic approaches combining a variety of diverse and contrasting disciplines are needed.The EU-ROS consortium (COST Action BM1203) was supported by the European Cooperation in Science and Technology (COST). The present overview represents the final Action dissemination summarizing the major achievements of COST Action BM1203 (EU-ROS) as well as research news and personal views of its members. Some authors were also supported by COST Actions BM1005 (ENOG) and BM1307 (PROTEOSTASIS), as well as funding from the European Commission FP7 and H2020 programmes, and several national funding agencies

    Epithelial–Mesenchymal Transition in Acute Leukemias

    No full text
    Epithelial–mesenchymal transition (EMT) is a metabolic process that confers phenotypic flexibility to cells and the ability to adapt to new functions. This transition is critical during embryogenesis and is required for the differentiation of many tissues and organs. EMT can also be induced in advanced-stage cancers, leading to further malignant behavior and chemotherapy resistance, resulting in an unfavorable prognosis for patients. Although EMT was long considered and studied only in solid tumors, it has been shown to be involved in the pathogenesis of hematological malignancies, including acute leukemias. Indeed, there is increasing evidence that EMT promotes the progression of acute leukemias, leading to the emergence of a more aggressive phenotype of the disease, and also causes chemotherapy resistance. The current literature suggests that the levels and activities of EMT inducers and markers can be used to predict prognosis, and that targeting EMT in addition to conventional therapies may increase treatment success in acute leukemias

    Aldehyde Dehydrogenase Genes as Prospective Actionable Targets in Acute Myeloid Leukemia

    No full text
    It has been previously shown that the aldehyde dehydrogenase (ALDH) family member ALDH1A1 has a significant association with acute myeloid leukemia (AML) patient risk group classification and that AML cells lacking ALDH1A1 expression can be readily killed via chemotherapy. In the past, however, a redundancy between the activities of subgroup members of the ALDH family has hampered the search for conclusive evidence to address the role of specific ALDH genes. Here, we describe the bioinformatics evaluation of all nineteen member genes of the ALDH family as prospective actionable targets for the development of methods aimed to improve AML treatment. We implicate ALDH1A1 in the development of recurrent AML, and we show that from the nineteen members of the ALDH family, ALDH1A1 and ALDH2 have the strongest association with AML patient risk group classification. Furthermore, we discover that the sum of the expression values for RNA from the genes, ALDH1A1 and ALDH2, has a stronger association with AML patient risk group classification and survival than either one gene alone does. In conclusion, we identify ALDH1A1 and ALDH2 as prospective actionable targets for the treatment of AML in high-risk patients. Substances that inhibit both enzymatic activities constitute potentially effective pharmaceutics

    Androgen regulated HN1 leads proteosomal degradation of androgen receptor (AR) and negatively influences AR mediated transactivation in prostate cells

    No full text
    WOS: 000301610000012PubMed ID: 22155408We recently reported that hematological and neurological expressed 1 (HN1) is a ubiquitously expressed, EGF-regulated gene. Expression of HN1 in prostate cell lines down-regulates PI3 K-dependent Akt activation. Here, we investigate whether HN1 is regulated by androgens through the putative androgen response elements (AREs) found in its promoter. Knockdown of HN1 expression by siRNA silencing leads to an increase in Akt((S473)) phosphorylation, resulting in the translocation of androgen receptor (AR) to the nucleus: these effects can be abrogated by the non-specific Akt inhibitor LY294002 but not by the ERK inhibitor PD98059. Furthermore, HN1 overexpression correlates with an increase in ubiquitination-mediated degradation (a consequence of the decrease in S213/210 phosphorylation of AR), ultimately resulting in the down-regulation of AR-mediated expression of the KLK3, KLK4, NKX.1 and STAMP2 genes. We also found that HN1 overexpression suppresses colony formation as well as R1881-mediated growth in LNCaP cells, while it has the opposite effect (increasing colony formation but not proliferation) in PC-3 and DU145 cells. Therefore, we suggest that HN1 maintains a balance between the androgen-regulated nuclear translocation of AR and steady-state Akt phosphorylation, predominantly in the absence of androgens. If so, the balance between cell growth and EGF- and AR-signaling must be tightly regulated by HN1. This work has important implications for prostate cancer research, as AR, EGFR and HN1 are known to be highly expressed in prostate adenocarcinomas. (C) 2011 Elsevier Ireland Ltd. All rights reserved.TUBITAKTurkiye Bilimsel ve Teknolojik Arastirma Kurumu (TUBITAK) [108S288 (COST_CANGENIN BM0703), 110S134]We would like to thank Prof. Fahri Saatcioglu, Ph.D. (University of Oslo, Institute of Biosciences, Oslo, Norway) for providing the pSG5-AR construct, and the rabbit polyclonal alpha-HN1, and alpha-NKX3.1 antibodies, and FABAL (Ege University, Faculty of Pharmacy, Izmir, Turkey) for technical support in the use of the xCELLigence real-time cell analysis system. This research was supported by Grants 108S288 (COST_CANGENIN BM0703) and 110S134 from TUBITAK to K.S.K

    NKX3.1 contributes to S phase entry and regulates DNA damage response (DDR) in prostate cancer cell lines

    Get PDF
    WOS: 000296215200023PubMed ID: 21945437NKX3.1 is an androgen-regulated homeobox gene that encodes a tissue-restricted transcription factor, which plays an important role in the differentiation of the prostate epithelium. Thus, the role of NKX3.1 as a functional topoisomerase I activity enhancer in cell cycle regulation and the DNA damage response (DDR) was explored in prostate cancer cell lines. As an early response to DNA damage following CPT-11 treatment, we found that there was an increase in the gamma H2AX((S139)) foci number and that total phosphorylation levels were reduced in PC-3 cells following ectopic NKX3.1 expression as well as in LNCaP cells following androgen administration. Furthermore, upon drug treatment, the increase in ATM((S1981)) phosphorylation was reduced in the presence of NKX3.1 expression, whereas DNA-PKcs expression was increased. Additionally, phosphorylation of CHK2((T68)) and NBS1((S343)) was abrogated by ectopic NKX3.1 expression, compared with the increasing levels in control PC-3 cells in a time-course experiment. Finally, NKX3.1 expression maintained a high cyclin D1 expression level regardless of drug treatment, while total gamma H2AX((S139)) phosphorylation remained depleted in PC-3, as well as in LNCaP, cells. Thus, we suggest that androgen regulated NKX3.1 maintains an active DDR at the intra S progression and contributes to the chemotherapeutic resistance of prostate cancer cells to DNA damaging compounds. (C) 2011 Elsevier Inc. All rights reserved.Turkish Scientific and Technological Research Council (COST action BM0703 CANGENIN)Turkiye Bilimsel ve Teknolojik Arastirma Kurumu (TUBITAK) [TUBITAK-106S200, -110S134, TUBITAK -108S288]; Ege UniversityEge University [06MUH004, 10MUH006]We would like to thank Dr. Timur Kose (Ege University, Department of Biostatistics and Biometry) for his kind assistance with the statistical analysis, Dr. Fahri Saatcioglu (University of Oslo, Oslo, Norway) for providing constructs, deletion mutants and the rabbit polyclonal alpha-NKX3.1 antibody, and Dr. Buket Kosova (Ege University, Izmir, Turkey) for the B-actin antibody. This research was supported with grants (TUBITAK-106S200, -110S134, COST action BM0703 CANGENIN (TUBITAK -108S288)) from the Turkish Scientific and Technological Research Council and BAP projects (06MUH004 and 10MUH006) by the Ege University to KSK

    HN1 interacts with gamma-tubulin to regulate centrosomes in advanced prostate cancer cells

    No full text
    Prostate cancer is one of the most common cancer for men worldwide with advanced forms showing supernumerary or clustered centrosomes. Hematological and neurological expressed 1 (HN1) also known as Jupiter Microtubule Associated Homolog 1 (JPT1) belongs to a small poorly understood family of genes that are evolutionarily conserved across vertebrate species. The co-expression network of HN1 from the TCGA PRAD dataset indicates the putative role of HN1 in centrosome-related processes in the context of prostate cancer. HN1 expression is low in normal RWPE-1 cells as compared to cancerous androgen-responsive LNCaP and androgen insensitive PC-3 cells. HN1 overexpression resulted in differential response for cell proliferation and cell cycle changes in RWPE-1, LNCaP, and PC-3 cells. Since HN1 overexpression increased the proliferation rate in PC-3 cells, these cells were used for functional characterization of HN1 in advanced prostate carcinogenesis. Furthermore, alterations in HN expression led to an increase in abnormal to normal nuclei ratio and increased chromosomal aberrations in PC-3 cells. We observed the co-localization of HN1 with gamma-tubulin foci in prostate cancer cells, further validated by immunoprecipitation. HN1 was observed as physically associated with gamma-tubulin and its depletion led to increased gamma-tubulin foci and disruption in microtubule spindle assembly. Higher HN1 expression was correlated with prostate cancer as compared to normal tissues. The restoration of HN1 expression after silencing suggested that it has a role in centrosome clustering, implicating a potential role of HN1 in cell division as well as in prostate carcinogenesis warranting further studies
    corecore