GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cells, MDPI AG, Vol. 11, No. 11 ( 2022-05-24), p. 1731-
    Abstract: Diseases that affect the mitochondrial electron transport chain (ETC) often manifest as threshold effect disorders, meaning patients only become symptomatic once a certain level of ETC dysfunction is reached. Cells can invoke mechanisms to circumvent reaching their critical ETC threshold, but it is an ongoing challenge to identify such processes. In the nematode Caenorhabditis elegans, severe reduction of mitochondrial ETC activity shortens life, but mild reduction actually extends it, providing an opportunity to identify threshold circumvention mechanisms. Here, we show that removal of ATL-1, but not ATM-1, worm orthologs of ATR and ATM, respectively, key nuclear DNA damage checkpoint proteins in human cells, unexpectedly lessens the severity of ETC dysfunction. Multiple genetic and biochemical tests show no evidence for increased mutation or DNA breakage in animals exposed to ETC disruption. Reduced ETC function instead alters nucleotide ratios within both the ribo- and deoxyribo-nucleotide pools, and causes stalling of RNA polymerase, which is also known to activate ATR. Unexpectedly, atl-1 mutants confronted with mitochondrial ETC disruption maintain normal levels of oxygen consumption, and have an increased abundance of translating ribosomes. This suggests checkpoint signaling by ATL-1 normally dampens cytoplasmic translation. Taken together, our data suggest a model whereby ETC insufficiency in C. elegans results in nucleotide imbalances leading to the stalling of RNA polymerase, activation of ATL-1, dampening of global translation, and magnification of ETC dysfunction. The loss of ATL-1 effectively reverses the severity of ETC disruption so that animals become phenotypically closer to wild type.
    Type of Medium: Online Resource
    ISSN: 2073-4409
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2661518-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Wiley ; 2010
    In:  Developmental Disabilities Research Reviews Vol. 16, No. 2 ( 2010-06), p. 200-218
    In: Developmental Disabilities Research Reviews, Wiley, Vol. 16, No. 2 ( 2010-06), p. 200-218
    Abstract: The extensive conservation of mitochondrial structure, composition, and function across evolution offers a unique opportunity to expand our understanding of human mitochondrial biology and disease. By investigating the biology of much simpler model organisms, it is often possible to answer questions that are unreachable at the clinical level. Here, we review the relative utility of four different model organisms, namely the bacterium Escherichia coli , the yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans , and the fruit fly Drosophila melanogaster , in studying the role of mitochondrial proteins relevant to human disease. E. coli are single cell, prokaryotic bacteria that have proven to be a useful model system in which to investigate mitochondrial respiratory chain protein structure and function. S. cerevisiae is a single‐celled eukaryote that can grow equally well by mitochondrial‐dependent respiration or by ethanol fermentation, a property that has proven to be a veritable boon for investigating mitochondrial functionality. C. elegans is a multicellular, microscopic worm that is organized into five major tissues and has proven to be a robust model animal for in vitro and in vivo studies of primary respiratory chain dysfunction and its potential therapies in humans. Studied for over a century, D. melanogaster is a classic metazoan model system offering an abundance of genetic tools and reagents that facilitates investigations of mitochondrial biology using both forward and reverse genetics. The respective strengths and limitations of each species relative to mitochondrial studies are explored. In addition, an overview is provided of major discoveries made in mitochondrial biology in each of these four model systems. © 2010 Wiley‐Liss, Inc. Dev Disabil Res Rev 2010;16:200–218.
    Type of Medium: Online Resource
    ISSN: 1940-5510 , 1940-5529
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2010
    detail.hit.zdb_id: 2423580-5
    SSG: 5,2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Impact Journals, LLC ; 2017
    In:  Oncotarget Vol. 8, No. 41 ( 2017-09-19), p. 69622-69640
    In: Oncotarget, Impact Journals, LLC, Vol. 8, No. 41 ( 2017-09-19), p. 69622-69640
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 3, No. Supplement_1 ( 2019-04-15)
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2019
    detail.hit.zdb_id: 2881023-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1971-1971
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1971-1971
    Abstract: The ETS transcription factor ESE-1 belongs to the top 5% of all overexpressed genes in ductal and lobular breast carcinomas. We have previously established that cytoplasmic expression of ESE-1 initiates transformation in benign MCF10A and MCF12A mammary epithelial cells, while nuclear ESE-1 maintains the transformed phenotype in luminal MCF7 and ZR-75-1 breast cancer cells. ESE-1 is also a transcriptional regulator of HER2 gene expression, and is highly expressed in HER2+ BT474 and SKBR3 cells. In this study we have delineated how ESE-1 controls transformation properties in HER2+ breast cancer cells, and have validated the clinical significance of ESE-1 overexpression in breast cancer patients. Here we show that ESE-1 not only maintains transformation properties in HER2-dependent, Herceptin (anti-HER2) sensitive HER2+ breast cancer cell lines BT474 and SKBR3, but also in HER2-independent, Herceptin-resistant clones of BT474 and SKBR3 cells, named BT474/HR20 and SKBR3/pool2 respectively. Using crystal violet growth assays, BrdU labeling, 2D clonogenic assays, and 3D colony formation in soft agar we show that knocking down ESE-1 inhibits proliferation, clonogenicity, and anchorage independent growth in SKBR3, BT474, BT474/HR20, and SKBR3/pool2 cell lines in-vitro. Mechanistically, ESE-1 knocked down Herceptin sensitive cells evinced a delay in cell cycle progression through G1, along with down-regulation of cyclin D1. Long-term stable inhibition of ESE-1 in these cells resulted in down-regulation of pAKT, which was not always correlated with inhibition of HER2 and pHER2. In the HER2-independent BT474/HR20 and SKBR3/pool2 cells, stable ESE-1 knockdown inhibited pAKT independent of HER2 down-regulation. Evaluation of the clinical significance of ESE-1 expression was done by IHC analysis on 319 patient tumor samples. While ≥ 20% nuclear ESE-1 expression associated with aggressive LN metastasis, ≥30% of cytoplasmic ESE-1 associated with poor prognosis in LN+ breast cancer patients. High cytoplasmic ESE-1 significantly associated with 27% of ER-negative, 35% of PR-negative, and 25% of HER2+ metastatic tumors. Log-rank analysis of disease free survival (DFS) in LN+ patients revealed that patients with ≥ 30% cytoplasmic ESE-1 had half the length of median DFS compared to LN+ patients with ≤ 30% cytoplasmic ESE-1 (p = 0.027). Analysis in all patients irrespective of their prognostic status depicted that patients expressing ≥ 20% nuclear ESE-1 or ≥ 30% cytoplasmic ESE-1 associated with poor median DFS compared to patients with & lt; 20% nuclear ESE-1 or & lt; 30% cytoplasmic ESE-1. Overall we established that ESE-1 is critical in both HER2-dependent, and HER2-independent tumorigenesis and provided novel mechanistic insights into ESE-1's mode of transformation. Finally, the data warrants a global role for ESE-1 as a prognostic marker in all breast cancer patients. Citation Format: Adwitiya Kar, Susan Edgerton, Ann Thor, Arthur Gutierrez-Hartmann. ESE-1 controls transformation properties in HER2+ breast cancer cells, and predicts poor prognostic status and survival in breast cancer patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1971. doi:10.1158/1538-7445.AM2015-1971
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2020
    In:  Current Opinion in Endocrinology, Diabetes & Obesity Vol. 27, No. 3 ( 2020-06), p. 170-176
    In: Current Opinion in Endocrinology, Diabetes & Obesity, Ovid Technologies (Wolters Kluwer Health), Vol. 27, No. 3 ( 2020-06), p. 170-176
    Abstract: The aim of this review is to summarize recent advances on development of in vivo preclinical models of adrenocortical carcinoma (ACC). Recent findings Significant progress has been achieved in the underlying molecular mechanisms of adrenocortical tumorigenesis over the last decade, and recent comprehensive profiling analysis of ACC tumors identified several genetic and molecular drivers of this disease. Therapeutic breakthroughs, however, have been limited because of the lack of preclinical models recapitulating the molecular features and heterogeneity of the tumors. Recent publications on genetically engineered mouse models and development of patient-derived ACC xenografts in both nude mice and humanized mice now provide researchers with novel tools to explore therapeutic targets in the context of heterogeneity and tumor microenvironment in human ACC. Summary We review current in-vivo models of ACC and discuss potential therapeutic opportunities that have emerged from these studies.
    Type of Medium: Online Resource
    ISSN: 1752-296X , 1752-2978
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2020
    detail.hit.zdb_id: 2273420-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2020
    In:  Breast Cancer Research and Treatment Vol. 182, No. 3 ( 2020-08), p. 601-612
    In: Breast Cancer Research and Treatment, Springer Science and Business Media LLC, Vol. 182, No. 3 ( 2020-08), p. 601-612
    Type of Medium: Online Resource
    ISSN: 0167-6806 , 1573-7217
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2004077-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Public Library of Science (PLoS) ; 2016
    In:  PLOS ONE Vol. 11, No. 9 ( 2016-9-7), p. e0162165-
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of the Endocrine Society, The Endocrine Society, Vol. 6, No. Supplement_1 ( 2022-11-01), p. A82-A82
    Abstract: Adrenocortical carcinoma (ACC) is an aggressive cancer with a significant risk of recurrence and high mortality rates. ACC has limited response to currently used treatments and there is an unmet clinical need to identify new treatment options. Disease rarity coupled with absence of preclinical models of disease heterogeneity contributed to limited therapeutic progress in ACC. We previously established new in vitro and in vivo ACC models (CUACC1, CUACC2, CUACC9) with variable genomic alterations commonly seen in ACC tumors. Prior work demonstrated that maternal leucine embryonic kinase (MELK) is an oncogenic kinase in ACC, and that it can be targeted with the MELK inhibitor OTSSP167. To further evaluate the anti-tumor efficacy and mode of action of OTSSP167, this study used genetically diverse ACC cell lines (CUACC1, CUACC2, H295R), patient derived xenografts (PDX) (CUACC1 and CUACC9) and a cell line xenograft model (H295R) of ACC. ACC PDXs treated with 10 mg/kg OTSSP167 had significantly reduced MELK expression, tumor growth inhibition of more than 80%, reduced mitosis, and increased tumor tissue necrosis. Transcriptomic and phospho-proteomic analysis at multiple treatment timepoints in in vitro models revealed that OTSSP167 induced wide transcriptomic and proteomic changes involving effectors of signal transduction, cell cycle, protein translation, and epigenetic regulation to exert its anti-tumorigenic effects. Evaluation of OTSSP167's role in protein synthesis via a puromycin based SUnSET (surface sensing of translation) and kinase assay established that OTSSP167 inhibits de-novo protein synthesis, and targets RSK1 and its substrate rpS6 (S235/S236). Varying the degree of MELK silencing in H295R cell lines using a doxycycline inducible shRNA construct, inhibited RSK1 phosphorylation at T573 and S380 indicating a MELK-RSK1 axis in ACC. Additionally, OTSSP167's effect on the DNA damage pathway was validated, which activated the G2/M checkpoint protein Wee1, increased inhibitory phosphorylation of CDK1, causing an accumulation of cyclin B1 and G2/M arrest independent of TP53 status. Combination treatment with the Wee1 inhibitor AZD1775 and OTSSP167 enhanced OTSSP167 mediated cytotoxic effects in in-vitro models with an increase in mitotic entry and caspase mediated apoptosis. In summary, OTSSP167 exerted its anti-tumorigenic effects via inhibition of the protein translation machinery, DNA damage and cell cycle dysregulation causing drug sensitization. Results from these studies are a first demonstration of the anti-cancer effects of OTSSP167 in sensitization to Wee1 inhibition in ACC. These data suggest that OTSSP167 is a potential therapeutic option for patients with ACC and could be combined with Wee1 inhibitors in a clinical trial using a synthetic lethal approach. Presentation: Saturday, June 11, 2022 12:45 p.m. - 1:00 p.m.
    Type of Medium: Online Resource
    ISSN: 2472-1972
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2022
    detail.hit.zdb_id: 2881023-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Endocrinology, The Endocrine Society, Vol. 159, No. 7 ( 2018-07-01), p. 2532-2544
    Abstract: Adrenocortical carcinoma (ACC) is an aggressive cancer with a 5-year survival rate 〈 35%. Mortality remains high due to lack of targeted therapies. Using bioinformatic analyses, we identified maternal embryonic leucine zipper kinase (MELK) as 4.1-fold overexpressed in ACC compared with normal adrenal samples. High MELK expression in human tumors correlated with shorter survival and with increased expression of genes involved in cell division and growth. We investigated the functional effects of MELK inhibition using newly developed ACC cell lines with variable MELK expression, CU-ACC1 and CU-ACC2, compared with H295R cells. In vitro treatment with the MELK inhibitor, OTSSP167, resulted in a dose-dependent decrease in rates of cell proliferation, colony formation, and cell survival, with relative sensitivity of each ACC cell line based upon the level of MELK overexpression. To confirm a MELK-specific antitumorigenic effect, MELK was inhibited in H295R cells via multiple short hairpin RNAs. MELK silencing resulted in 1.9-fold decrease in proliferation, and 3- to 10-fold decrease in colony formation in soft agar and clonogenicity assays, respectively. In addition, although MELK silencing had no effect on survival in normoxia, exposure to a hypoxia resulted in a sixfold and eightfold increase in apoptosis as assessed by caspase-3 activation and TUNEL, respectively. Together these data suggest that MELK is a modulator of tumor cell growth and survival in a hypoxic microenvironment in adrenal cancer cells and support future investigation of its role as a therapeutic kinase target in patients with ACC.
    Type of Medium: Online Resource
    ISSN: 1945-7170
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2018
    detail.hit.zdb_id: 2011695-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...