Skip to main content
Erschienen in: Molecular Cancer 1/2013

Open Access 01.12.2013 | Short communication

Phospholipid Scramblase 1, an interferon-regulated gene located at 3q23, is regulated by SnoN/SkiL in ovarian cancer cells

verfasst von: Karthik M. Kodigepalli, Pavana Anur, Paul Spellman, Peter J. Sims, Meera Nanjundan

Erschienen in: Molecular Cancer | Ausgabe 1/2013

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Treatment of advanced stage ovarian cancer continues to be challenging due to acquired drug resistance and lack of early stage biomarkers. Genes identified to be aberrantly expressed at the 3q26.2 locus (i.e. SnoN/SkiL) have been implicated in ovarian cancer pathophysiology. We have previously shown that SnoN expression is increased in advanced stage ovarian cancers and alters cellular response to arsenic trioxide (As2O3).

Findings

We now demonstrate increased DNA copy number levels (TCGA data) of phospholipid scramblase 1 (PLSCR1, located at 3q23) whose transcript expression in ovarian cell lines is highly correlated with SnoN mRNA. Interestingly, SnoN can modulate PLSCR1 mRNA levels in the absence/presence of interferon (IFN-2α). Both IFN-2α and As2O3 treatment can modulate PLSCR1 mRNA levels in ovarian carcinoma cells. However, SnoN siRNA does not lead to altered PLSCR1 protein implicating other events needed to modulate its protein levels. In addition, we report that PLSCR1 can modulate aspects of the As2O3 cellular response.

Conclusions

Our findings warrant further investigation into the role of PLSCR1 in ovarian cancer development and chemoresistance.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-12-32) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MN conceived and supervised the study. PJS developed the following constructs for use in this work: pGL3-PLSCR1 promoter, wild type PLSCR1 in pcDNA3.1, and C/A PLSCR1 mutant in pcDNA3.1. KMK, PA, PS, and MN performed the research and analyzed the data. KMK and MN co-wrote the paper and all authors approved the final manuscript.
Abkürzungen
PLSCR1
Phospholipid Scramblase 1
SnoN/SkiL
Ski Related Novel Protein N
EVI1
Ecotropic Viral Integration Site-1
PIK3CA
phosphatidylinositol-3-kinase catalytic subunit-α
PKCι
Protein Kinase C iota
aCGH
Array Comparative Genomic Hybridization
TCGA
The Cancer Genome Atlas
PAI-1
Plasminogen Activator Inhibitor-1
TGFβ
Transforming Growth Factor-β
As2O3
Arsenic trioxide
CHX
Cycloheximide
IFN
Interferon
ROS
Reactive Oxygen Species
NAC
N-Acetyl-L-Cysteine
LC3
Microtubule-associated protein light chain 3
GFP
Green fluorescent protein
PI
Propidium Iodide
PARP
Poly-ADP Ribose Polymerase
T80
Immortalized (LTAg/hTERT) normal ovarian surface epithelial cells
APL
Acute Promyelocytic Leukemia.

Findings

Epithelial ovarian cancer represents the most common gynecological cancer in women with an unfortunate high mortality rate due to acquired chemotherapeutic resistance [1]. Our earlier published studies indicate that the 3q26.2 chromosomal region is highly amplified in ovarian cancers [2] and harbors various oncogenes including EVI1 [2], PKCι [3], and SnoN/SkiL [4]. In particular, we previously demonstrated that SnoN, a negative transcriptional regulator of TGFβ signaling, modulates the pro-survival autophagic pathway in response to arsenic trioxide (As2O3), a chemotherapeutic agent used in the treatment of acute promyelocytic leukemia (APL) [5]. Interestingly, there are reports which indicate that genes located at and proximal to the 3q26 locus may regulate each other. For instance, both EVI1 and PIK3CA can regulate SnoN expression [6, 7]. Herein, we now report that the expression of phospholipid scramblase 1 (PLSCR1), located at 3q23, can be modulated via SnoN. PLSCR1 has been implicated in maintaining plasma membrane lipid asymmetry, regulating growth factor signaling pathways, in modulating tumor growth in mouse xenograft models [8], and cancer development [9, 10]. The role of PLSCR1 in ovarian cancer and in modulating response to chemotherapeutic agents has yet to be fully understood.
Our previous aCGH studies from 235 ovarian cancer patient samples demonstrated that SnoN was increased at the DNA copy number level [4]. We now identify through Oncomine bioinformatic analyses (ovarian TCGA dataset (https://​tcga-data.​nci.​nih.​gov.​tcga/​) that the DNA copy number levels of PLSCR1 in addition to SnoN are altered similarly (Figure 1A). Furthermore, using cBioportal [11], we identified that SnoN is amplified in 31% of the cases whereas PLSCR1 is amplified in 13% of the cases (70 out of 570 samples amplified both genes). To determine whether SnoN and PLSCR1 genes are co-amplified, we performed linear regression on copy number variation (CNV) estimates (Additional file 1: Methods and Materials) for SnoN and PLSCR1 genes in R (http://​www.​R-project.​org/​) (Figure 1B). In ovarian cancers with PLSCR1 amplification, SnoN is gained. When SnoN is amplified, PLSCR1 is only gained in ~33% of the samples (R2 = 0.2474) (Figure 1B). We next evaluated the RNA and protein levels of PLSCR1 in various normal and malignant ovarian cell lines via real-time PCR and western analysis. Similar to SnoN, PLSCR1 expression was low in normal immortalized T80 ovarian cells and highly expressed in the ovarian cancer cell lines (Figure 1C and E). Although PLSCR1 and SnoN expression were highly correlated (via linear regression) at the RNA level (Figure 1D), there was a discordance at the protein level (Figure 1F) which has been reported previously for other genes [12, 13]. Furthermore, the DNA copy number of PLSCR1 and SnoN is nearly always the same in ovarian cancer cell lines (R2 = 0.6411) (Additional file 2: Table S1). Collectively, these results demonstrate that, PLSCR1 is increased at the DNA and RNA levels in ovarian cancers and cell lines in comparison to normal cells, similar to SnoN, and can be co-amplified in a certain proportion of ovarian cancer specimens. However, there likely exist additional levels of regulation which contribute to modulating PLSCR1 protein levels.
Since PLSCR1 is located in close proximity to SnoN at the 3q locus [2], we next assessed whether SnoN could modulate PLSCR1 expression. To address this question, we reduced SnoN expression via siRNA in HEY ovarian carcinoma cells (cell line used previously to investigate role of SnoN [5] and PLSCR1 [8]); this was followed by quantitation of PLSCR1 mRNA levels via real-time PCR. Upon SnoN knockdown (~88% and 95% at RNA and protein level, respectively), we observed a significant reduction (~35%) in PLSCR1 mRNA (Figure 2A) implicating SnoN in the regulation of PLSCR1 transcription. These results were validated by utilizing the PLSCR1 promoter upon SnoN knockdown in T80 cells (Figure 2B) or with TGFβ (50 pM) (Figure 2C); both conditions led to a marked reduction in PLSCR1 promoter activity suggesting that activation of the TGFβ signaling cascade downregulates PLSCR1 expression. PLSCR1 mRNA levels were also down-regulated following 24 h TGFβ treatment (Figure 2D, left panel). SnoN mRNA has been previously shown to increase 1–3 hours post-TGFβ treatment [4] (Figure 2D, right panel) implicating discordance between SnoN and PLSCR1 mRNA levels with TGFβ. Intriguingly, overexpression of both the wild type and C/A PLSCR1 mutant (which localizes to the nuclear compartment [14]) in T80 cells led to a marked induction of plasminogen activator inhibitor-1 (PAI-1) expression (Figure 2E); these results suggest that PLSCR1 could modulate TGFβ cellular responses, similar to SnoN [4].
Since PLSCR1 is an interferon (IFN)-inducible gene [15, 16], we next determined whether SnoN could, in part, modulate PLSCR1 expression upon IFN-2α treatment. Supporting previous reports, treatment of HEY cells with IFN-2α (3000 IU/ml) led to a dramatic increase in PLSCR1 protein from 6 up to 24 hours (Figure 3A, 3.7-fold) and RNA (Figure 3B, 2.9-fold). Similar results were also observed in a series of IFN-resistant and sensitive pancreatic cancer cell lines (Additional file 3: Figure S1, A-F). Strikingly, SnoN protein was induced (in the absence of SnoN mRNA changes (Additional file 4: Figure S2, A)) at 3 h post-IFN treatment; changes in SnoN occurred prior to those observed in PLSCR1. These results implicate SnoN in the transcriptional regulation of PLSCR1 expression upon IFN treatment. In addition, we noted that the induced PLSCR1 localized predominantly at the plasma membrane in HEY cells (Figure 3C and D), assessed via immunofluorescence and subcellular fractionation (with a very minor fraction localizing to the nuclear compartment). Since chemotherapeutic agents can generate intracellular reactive oxygen species (ROS) which modulates expression levels of various proteins [5], we next assessed whether the changes we observed in PLSCR1 and SnoN expression with IFN were due to ROS. Thus, we co-treated HEY cells with N-acetyl-L-cysteine (NAC), an anti-oxidant free radical scavenger, together with IFN for 9 h. However, there were no marked changes in PLSCR1 and SnoN protein levels in the presence of NAC (Additional file 4: Figure S2, B); these results suggest that the IFN-induced changes in SnoN and PLSCR1 may be independent of ROS. Strikingly, knockdown of SnoN levels (via siRNA) in HEY cells in the presence of IFN-2α (6 h, 3000 IU/ml) not only effectively reduced SnoN levels but also PLSCR1 RNA (Figure 3E and F, 1.8-fold). However, changes in PLSCR1 protein were again not detected with IFN-treatment following SnoN siRNA (Additional file 4: Figure S2, C); we propose that this could be due to the long half-life of PLSCR1 protein (assessed utilizing cycloheximide (CHX), an inhibitor of protein translation (results not shown)) or additional mechanisms needed to contribute with SnoN to modulate PLSCR1 protein.
We have previously demonstrated the role of As2O3 as an effective chemotherapeutic agent inducing cell death in ovarian cancer cells, antagonized by autophagy mediated by SnoN induction [5]. We first assessed whether PLSCR1 protein is altered upon As2O3 treatment in ovarian cancer cells. In this regard, we treated HEY cells with 25 μM As2O3 (0 – 24 h). In contrast to SnoN (increasing between 6 – 24 h), we noted a marked reduction in PLSCR1 protein (~75% reduction) (Figure 4A and B). We next determined whether this reduction in PLSCR1 protein was due to proteasomal degradation via the use of MG132 (proteasome inhibitor). Co-treatment of HEY cells with 5 and 25 μM As2O3 for 6 h and 18 h with 5 μM MG132 did not lead to a significant recovery in PLSCR1 levels; these results suggest a mechanism of PLSCR1 protein regulation independent of the proteasome (Figure 4C). Indeed, As2O3 also alters PLSCR1 RNA levels which might together reflect As2O3-induced transcriptional regulation of PLSCR1 (Figure 4D). In order to determine whether PLSCR1 plays a role in modulating As2O3–induced cell death response in ovarian cancer cells, we reduced PLSCR1 expression via siRNA. Upon knockdown of PLSCR1 in the presence of As2O3, we observed a marked increase in the levels of caspase-3 activity (results not shown) as well as cleaved PARP ((7.9-fold) a marker of apoptosis, Figure 4E) concurrent with reduction in LC3-II ((2.5-fold), a marker of autophagy, Figure 4E) validated by GFP-LC3 autophagy assays ((~20%), Figure 4F). Similar to SnoN [5], these results suggest that PLSCR1 may contribute to the As2O3-induced apoptotic and autophagic response.
In the current study, we demonstrate that PLSCR1 and SnoN DNA copy number as well as their RNA levels are correlated. By modulating SnoN expression, PLSCR1 mRNA levels appear to be co-regulated (Figure 4G). Of interest, SnoN knockdown does not alter PLSCR1 protein possibly suggesting that other mediators are involved in its regulation. Nonetheless, similar to SnoN, reduction in PLSCR1 levels appears to increase the cellular sensitivity to As2O3. Whether PLSCR1 modulates sensitivity to carboplatin/paclitaxel or whether the effects of As2O3 and TGFβ are mediated via IFN remain to be investigated. Thus, further investigations are warranted to delve into the significance of these findings in ovarian cancer development and chemoresistance.

Acknowledgements

This work was supported by funds from the National Institute of Health RO1 CA 123219 and University of South Florida Start-up Funds to Meera Nanjundan. This work was also supported in part by the Flow Cytometry Core Facility at the College of Medicine, University of South Florida. We thank Dawn Smith, Annemarie Boland, and Hussain Basrawala for their technical assistance with the studies presented herein. We also are grateful to Stephanie Rockfield and Katherine Allen for their assistance with figure preparation.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MN conceived and supervised the study. PJS developed the following constructs for use in this work: pGL3-PLSCR1 promoter, wild type PLSCR1 in pcDNA3.1, and C/A PLSCR1 mutant in pcDNA3.1. KMK, PA, PS, and MN performed the research and analyzed the data. KMK and MN co-wrote the paper and all authors approved the final manuscript.
Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Bast RC, Hennessy B, Mills GB: The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009, 9 (6): 415-428. 10.1038/nrc2644.PubMedCentralCrossRefPubMed Bast RC, Hennessy B, Mills GB: The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009, 9 (6): 415-428. 10.1038/nrc2644.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Nanjundan M, Nakayama Y, Cheng KW, Lahad J, Liu J, Lu K, Kuo WL, Smith-McCune K, Fishman D, Gray JW: Amplification of MDS1/EVI1 and EVI1, located in the 3q26.2 amplicon, is associated with favorable patient prognosis in ovarian cancer. Cancer Res. 2007, 67 (7): 3074-3084. 10.1158/0008-5472.CAN-06-2366.CrossRefPubMed Nanjundan M, Nakayama Y, Cheng KW, Lahad J, Liu J, Lu K, Kuo WL, Smith-McCune K, Fishman D, Gray JW: Amplification of MDS1/EVI1 and EVI1, located in the 3q26.2 amplicon, is associated with favorable patient prognosis in ovarian cancer. Cancer Res. 2007, 67 (7): 3074-3084. 10.1158/0008-5472.CAN-06-2366.CrossRefPubMed
3.
Zurück zum Zitat Eder AM, Sui X, Rosen DG, Nolden LK, Cheng KW, Lahad JP, Kango-Singh M, Lu KH, Warneke CL, Atkinson EN: Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci USA. 2005, 102 (35): 12519-12524. 10.1073/pnas.0505641102.PubMedCentralCrossRefPubMed Eder AM, Sui X, Rosen DG, Nolden LK, Cheng KW, Lahad JP, Kango-Singh M, Lu KH, Warneke CL, Atkinson EN: Atypical PKCiota contributes to poor prognosis through loss of apical-basal polarity and cyclin E overexpression in ovarian cancer. Proc Natl Acad Sci USA. 2005, 102 (35): 12519-12524. 10.1073/pnas.0505641102.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Nanjundan M, Cheng KW, Zhang F, Lahad J, Kuo WL, Schmandt R, Smith-McCune K, Fishman D, Gray JW, Mills GB: Overexpression of SnoN/SkiL, amplified at the 3q26.2 locus, in ovarian cancers: a role in ovarian pathogenesis. Mol Oncol. 2008, 2 (2): 164-181. 10.1016/j.molonc.2008.05.001.PubMedCentralCrossRefPubMed Nanjundan M, Cheng KW, Zhang F, Lahad J, Kuo WL, Schmandt R, Smith-McCune K, Fishman D, Gray JW, Mills GB: Overexpression of SnoN/SkiL, amplified at the 3q26.2 locus, in ovarian cancers: a role in ovarian pathogenesis. Mol Oncol. 2008, 2 (2): 164-181. 10.1016/j.molonc.2008.05.001.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Smith DM, Patel S, Raffoul F, Haller E, Mills GB, Nanjundan M: Arsenic trioxide induces a beclin-1-independent autophagic pathway via modulation of SnoN/SkiL expression in ovarian carcinoma cells. Cell Death Differ. 2010, 17 (12): 1867-1881. 10.1038/cdd.2010.53.PubMedCentralCrossRefPubMed Smith DM, Patel S, Raffoul F, Haller E, Mills GB, Nanjundan M: Arsenic trioxide induces a beclin-1-independent autophagic pathway via modulation of SnoN/SkiL expression in ovarian carcinoma cells. Cell Death Differ. 2010, 17 (12): 1867-1881. 10.1038/cdd.2010.53.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Yatsula B, Lin S, Read AJ, Poholek A, Yates K, Yue D, Hui P, Perkins AS: Identification of Binding Sites of EVI1 in Mammalian Cells. J Biol Chem. 2005, 280 (35): 30712-30722. 10.1074/jbc.M504293200.CrossRefPubMed Yatsula B, Lin S, Read AJ, Poholek A, Yates K, Yue D, Hui P, Perkins AS: Identification of Binding Sites of EVI1 in Mammalian Cells. J Biol Chem. 2005, 280 (35): 30712-30722. 10.1074/jbc.M504293200.CrossRefPubMed
7.
Zurück zum Zitat Kodigepalli KM, Dutta PS, Bauckman KA, Nanjundan M: SnoN/SkiL expression is modulated via arsenic trioxide-induced activation of the PI3K/AKT pathway in ovarian cancer cells. FEBS Lett. 2013, 587 (1): 5-16. 10.1016/j.febslet.2012.11.003.PubMedCentralCrossRefPubMed Kodigepalli KM, Dutta PS, Bauckman KA, Nanjundan M: SnoN/SkiL expression is modulated via arsenic trioxide-induced activation of the PI3K/AKT pathway in ovarian cancer cells. FEBS Lett. 2013, 587 (1): 5-16. 10.1016/j.febslet.2012.11.003.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Silverman RH, Halloum A, Zhou A, Dong B, Al-Zoghaibi F, Kushner D, Zhou Q, Zhao J, Wiedmer T, Sims PJ: Suppression of ovarian carcinoma cell growth in vivo by the interferon-inducible plasma membrane protein, phospholipid scramblase 1. Cancer Res. 2002, 62 (2): 397-402.PubMed Silverman RH, Halloum A, Zhou A, Dong B, Al-Zoghaibi F, Kushner D, Zhou Q, Zhao J, Wiedmer T, Sims PJ: Suppression of ovarian carcinoma cell growth in vivo by the interferon-inducible plasma membrane protein, phospholipid scramblase 1. Cancer Res. 2002, 62 (2): 397-402.PubMed
9.
Zurück zum Zitat Bateman A, Finn RD, Sims PJ, Wiedmer T, Biegert A, Soding J: Phospholipid scramblases and Tubby-like proteins belong to a new superfamily of membrane tethered transcription factors. Bioinformatics (Oxford, England). 2009, 25 (2): 159-162. 10.1093/bioinformatics/btn595.CrossRef Bateman A, Finn RD, Sims PJ, Wiedmer T, Biegert A, Soding J: Phospholipid scramblases and Tubby-like proteins belong to a new superfamily of membrane tethered transcription factors. Bioinformatics (Oxford, England). 2009, 25 (2): 159-162. 10.1093/bioinformatics/btn595.CrossRef
10.
Zurück zum Zitat Kuo YB, Chan CC, Chang CA, Fan CW, Hung RP, Hung YS, Chen KT, Yu JS, Chang YS, Chan EC: Identification of phospholipid scramblase 1 as a biomarker and determination of its prognostic value for colorectal cancer. Mol Med. 2011, 17 (1–2): 41-47.PubMedCentralPubMed Kuo YB, Chan CC, Chang CA, Fan CW, Hung RP, Hung YS, Chen KT, Yu JS, Chang YS, Chan EC: Identification of phospholipid scramblase 1 as a biomarker and determination of its prognostic value for colorectal cancer. Mol Med. 2011, 17 (1–2): 41-47.PubMedCentralPubMed
11.
Zurück zum Zitat Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E: The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discovery. 2012, 2 (5): 401-404. 10.1158/2159-8290.CD-12-0095.CrossRefPubMed Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E: The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discovery. 2012, 2 (5): 401-404. 10.1158/2159-8290.CD-12-0095.CrossRefPubMed
12.
Zurück zum Zitat Wahid F, Shehzad A, Khan T, Kim YY: MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta. 2010, 1803 (11): 1231-1243. 10.1016/j.bbamcr.2010.06.013.CrossRefPubMed Wahid F, Shehzad A, Khan T, Kim YY: MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochim Biophys Acta. 2010, 1803 (11): 1231-1243. 10.1016/j.bbamcr.2010.06.013.CrossRefPubMed
13.
Zurück zum Zitat Kortmann J, Sczodrok S, Rinnenthal J, Schwalbe H, Narberhaus F: Translation on demand by a simple RNA-based thermosensor. Nucleic Acids Res. 2011, 39 (7): 2855-2868. 10.1093/nar/gkq1252.PubMedCentralCrossRefPubMed Kortmann J, Sczodrok S, Rinnenthal J, Schwalbe H, Narberhaus F: Translation on demand by a simple RNA-based thermosensor. Nucleic Acids Res. 2011, 39 (7): 2855-2868. 10.1093/nar/gkq1252.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Wiedmer T, Zhao J, Nanjundan M, Sims PJ: Palmitoylation of phospholipid scramblase 1 controls its distribution between nucleus and plasma membrane. Biochemistry. 2003, 42 (5): 1227-1233. 10.1021/bi026679w.CrossRefPubMed Wiedmer T, Zhao J, Nanjundan M, Sims PJ: Palmitoylation of phospholipid scramblase 1 controls its distribution between nucleus and plasma membrane. Biochemistry. 2003, 42 (5): 1227-1233. 10.1021/bi026679w.CrossRefPubMed
15.
Zurück zum Zitat Der SD, Zhou A, Williams BR, Silverman RH: Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci USA. 1998, 95 (26): 15623-15628. 10.1073/pnas.95.26.15623.PubMedCentralCrossRefPubMed Der SD, Zhou A, Williams BR, Silverman RH: Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci USA. 1998, 95 (26): 15623-15628. 10.1073/pnas.95.26.15623.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Dong B, Zhou Q, Zhao J, Zhou A, Harty RN, Bose S, Banerjee A, Slee R, Guenther J, Williams BR: Phospholipid scramblase 1 potentiates the antiviral activity of interferon. J Virol. 2004, 78 (17): 8983-8993. 10.1128/JVI.78.17.8983-8993.2004.PubMedCentralCrossRefPubMed Dong B, Zhou Q, Zhao J, Zhou A, Harty RN, Bose S, Banerjee A, Slee R, Guenther J, Williams BR: Phospholipid scramblase 1 potentiates the antiviral activity of interferon. J Virol. 2004, 78 (17): 8983-8993. 10.1128/JVI.78.17.8983-8993.2004.PubMedCentralCrossRefPubMed
Metadaten
Titel
Phospholipid Scramblase 1, an interferon-regulated gene located at 3q23, is regulated by SnoN/SkiL in ovarian cancer cells
verfasst von
Karthik M. Kodigepalli
Pavana Anur
Paul Spellman
Peter J. Sims
Meera Nanjundan
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2013
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-12-32

Weitere Artikel der Ausgabe 1/2013

Molecular Cancer 1/2013 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.