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: Cell, Elsevier BV, Vol. 184, No. 13 ( 2021-06), p. 3376-3393.e17
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 187009-9
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2417-2417
    Abstract: High-grade serous ovarian cancers (HGSOCs) are the most common subtype of epithelial ovarian cancer. Pathways driving HGSOC development are poorly understood, and the cellular origins are debated in the literature. Previously, ovarian surface epithelial cells (OSECs) were thought to be cellular precursors of HGSOC, but there is now strong evidence that at least half of all HGSOCs arise from fallopian tube secretory epithelial cells (FTSECs). We took a large-scale integrated transcriptomic-epigenomic profiling approach to defining the molecular hallmarks of HGSOC and to explore the disease cell-of-origin. RNA sequencing was performed on 121 OSEC cultures and 84 FTSEC cultures and integrated with 394 HGSOC transcriptomes profiled by TCGA. We identified around 100 genes that differentiate OSECs and FTSECs, and pathway analysis of this gene list identified ovarian cancer genes as highly enriched (p=2.2x10-8), with ZEB1 and estrogen the most significant predicted upstream regulators (P & lt;3.7x10-7). We integrated gene expression data with clinical metadata to identify associations between gene expression signatures and clinical variables and found that in patients diagnosed with ovarian cancer, OSEC and FTSEC profiles converged, suggestive of a field cancerization phenotype occurring in the normal OSECs on the unaffected ovary contralateral to a clinically diagnosed ovarian carcinoma. Transcriptomic signatures for OSECs, FTSECs and HGSOCs were then merged with superenhancers defined by performing chromatin immunoprecipitation data for acetylated H3K27 profiled in primary HGSOC tissue specimens. We detected many more superenhancer-target gene relationships in the normal cells than in the tumor cells, suggesting superenhancer fingerprints define the cell of origin rather than determining upregulation of oncogenes driving cancer. To explore this further we determined the similarities of HGSOCs to OSEC and FTSEC using a machine learning model. Using a pre-defined gene signature of OSECs and FTSECs, our model (high specificity and sensitivity; 95% CI = 0.77-1, p-value=6.1-5) was applied to HGSOCs and we observed that the majority of HGSOCs were classified as FTSEC-like. Interestingly, the mesenchymal subtypes were enriched as FTSEC-like. Moreover, a subset of HGSOCs were characterized as OSEC-like, and these tumors were associated with poorer outcomes. Our data describe molecular subgroups within HGSOC precursor tissues as well as defining the transcriptional networks that define HGSOC precursor tissues, drive HGSOC development; furthermore we describe the interplay between the epigenetic landscape and the transcriptome in these cell types. In closing, these large-scale integrative analyses support a predominantly FTSEC origin for HGSOCs, but reinforce conclusions that OSECs cannot be excluded as origins of HGSOC, and may represent a subset of tumors with a different biology. Citation Format: Kate Lawrenson, Marcos Abraao, Felipe Segato, Janet M. Lee, Simon Coetzee, Ji-Heui Seo, Matthew L. Freedman, Dennis Hazelett, Simon Gayther, Houtan Noushmehr. Large scale integrated transcriptomic and epigenetic profiling defines the molecular hallmarks of HGSOC and disease origins [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2417. doi:10.1158/1538-7445.AM2017-2417
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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 ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 22_Supplement ( 2019-11-15), p. GMM-042-GMM-042
    Abstract: Ovarian cancer is a rare but highly lethal gynecologic malignancy, causing around 14,000 deaths annually in the US. The current standard of care for ovarian cancer includes a combination of surgical cytoreduction and platinum-based chemotherapy. Despite these aggressive treatments, most patients suffer from fatal relapses that indicate an urgent need for novel targeted therapies. Master Transcription Factors (MTFs) are transcriptional regulators responsible for cellular identity, and are associated with cell type specific regions of active chromatin termed super-enhancers (SEs). In cancer, MTFs are drivers of tumor development across a variety of organs, and MTF interfering therapies are one of the most successful therapies available today. To further identify tumor-specific MTFs, we have developed the ‘Cancer Core Transcription factor Specificity (CaCTS)' algorithm and prioritized MTFs from a pan-cancer cohort of 10,000 tumors profiled by RNA-sequencing. Results demonstrated that many tumor MTFs derived from CaCTS were specific to its normal tissue-type, validating the algorithm's ability to identify factors involved in cellular identity. On the other hand, a set of highly expressed TFs were frequently observed in multiple cancer types, indicating two distinct classes of MTFs: lineage-specific MTFs and pan-cancer MTFs. In High-Grade Serous Ovarian Cancer (HGSOC), CaCTS prioritized 21 TFs with lineage specific MTF traits. All 21 candidate MTFs were highly expressed in primary HGSOC specimens and aligned with HGSOC SEs identified via H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq). As expected, WT1 and PAX8 were identified, transcription factors previously implicated to be essential in HGSOC lineage specificity. Our algorithm also identified ZNF217, a novel candidate MTF in HGSOC. ZNF217 plays a key role in breast cancer, a tumor type that shares many epidemiological, hormonal and genetic risk factors with HGSOC. Mining of a publicly available gene knockout experiment in 13 HGSOC cell lines revealed ZNF217 to have similar levels of gene essentiality as PAX8. We are currently examining the role of ZNF217 as a MTF in HGSOC by determining its target genes, binding partners, and cellular consequences of siRNA-mediated knockdown. We expect ZNF217 to occupy SEs that drive expression of genes critical for cell identity, co-occupy SEs with other candidate MTFs, and depletion of ZNF217 to result in loss of cell state. Understanding the mechanisms of transcriptional control in HGSOC will ultimately help to identify clinically relevant drivers which can serve as novel therapeutic targets for this devastating disease. Citation Format: Robbin Nameki, Marcos A. S. Fonseca, Jessica Reddy, Kevin C. Vavra, Felipe Segato, Rosario I. Corona, Ji-Heui Seo, Annie Liu, Forough Abbasi, Xianzhi Lin, Beth Y. Karlan, Matthew L. Freedman, Simon A. Gayther, Houtan Noushmehr, Kate Lawrenson. PAN-CANCER TRANSCRIPTION FACTOR ANALYSES IDENTIFY ZNF217 AS A NOVEL DRIVER IN HGSOC [abstract]. In: Proceedings of the 12th Biennial Ovarian Cancer Research Symposium; Sep 13-15, 2018; Seattle, WA. Philadelphia (PA): AACR; Clin Cancer Res 2019;25(22 Suppl):Abstract nr GMM-042.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: JNCI: Journal of the National Cancer Institute, Oxford University Press (OUP), Vol. 114, No. 11 ( 2022-11-14), p. 1533-1544
    Abstract: Known risk alleles for epithelial ovarian cancer (EOC) account for approximately 40% of the heritability for EOC. Copy number variants (CNVs) have not been investigated as EOC risk alleles in a large population cohort. Methods Single nucleotide polymorphism array data from 13 071 EOC cases and 17 306 controls of White European ancestry were used to identify CNVs associated with EOC risk using a rare admixture maximum likelihood test for gene burden and a by-probe ratio test. We performed enrichment analysis of CNVs at known EOC risk loci and functional biofeatures in ovarian cancer–related cell types. Results We identified statistically significant risk associations with CNVs at known EOC risk genes; BRCA1 (PEOC = 1.60E-21; OREOC = 8.24), RAD51C (Phigh-grade serous ovarian cancer [HGSOC] = 5.5E-4; odds ratio [OR] HGSOC = 5.74 del), and BRCA2 (PHGSOC = 7.0E-4; ORHGSOC = 3.31 deletion). Four suggestive associations (P  & lt; .001) were identified for rare CNVs. Risk-associated CNVs were enriched (P  & lt; .05) at known EOC risk loci identified by genome-wide association study. Noncoding CNVs were enriched in active promoters and insulators in EOC-related cell types. Conclusions CNVs in BRCA1 have been previously reported in smaller studies, but their observed frequency in this large population-based cohort, along with the CNVs observed at BRCA2 and RAD51C gene loci in EOC cases, suggests that these CNVs are potentially pathogenic and may contribute to the spectrum of disease-causing mutations in these genes. CNVs are likely to occur in a wider set of susceptibility regions, with potential implications for clinical genetic testing and disease prevention.
    Type of Medium: Online Resource
    ISSN: 0027-8874 , 1460-2105
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2022
    detail.hit.zdb_id: 2992-0
    detail.hit.zdb_id: 1465951-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Nature Genetics, Springer Science and Business Media LLC, Vol. 51, No. 5 ( 2019-5), p. 815-823
    Type of Medium: Online Resource
    ISSN: 1061-4036 , 1546-1718
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 1494946-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1494-1494
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1494-1494
    Abstract: High-grade serous ovarian cancer (HGSOC) is the most common and lethal subtype of ovarian cancer. Recent data indicate that fallopian tube secretory epithelial cells (FTSECs) represent the cell of origin for HGSOC. While many studies have characterized molecular features associated with HGSOC biology, the master transcription factors (TFs) that drive disease development are not known. To identify master TFs for HGSOC, we first performed an integrative analysis of gene expression profiles from 73 FTSEC samples and 394 HGSOCs from The Cancer Genome Atlas (TCGA) project. We then integrated these data with chromatin immunoprecipitation sequencing (ChIP-seq) analyses performed on primary HGSOC samples to select active chromatin regions marked by H3k27ac and to identify super-enhancer (SE) regions. These analyses identified overexpressed TFs that coincide with tumor-specific SEs, a hallmark of master TFs. We identified multiple putative master TFs including SOX18, which was highly overexpressed in HGSOCs relative to normal FTSECs (log2 fold change in expression = 3.5, p = 2.5x10-19). SOX18 is a TF that regulates development of blood and lymphatic vessels, but its association with HGSOC development has not been reported before. We examined the expression of SOX18 in ~8,000 tumors representing 17 tumor types from TCGA. SOX18 expression was significantly elevated in HGSOC relative to any other tumor types, suggesting it is highly specific to this cancer. We found that SOX18 is highly expressed in the HGSOC cell lines UWB1.289, Kuramochi, and EFO21, and we then performed lentiviral-mediated shRNA knockdown of SOX18 in these cell lines to establish the effects of SOX18 depletion on neoplastic phenotypes. In ongoing experiments, we are performing gene expression profiling and H3k27ac ChIP-seq analysis after SOX18 knockdown to identify SOX18 target genes and characterize the landscape of SOX18 binding sites in HGSOC. These studies will likely identify novel molecular biomarkers that may represent much-needed therapeutic targets for HGSOC. Citation Format: Annie Y. Liu, Kevin C. Vavra, Rosario I. Corona, Forough Abassi, Marcos Fonseca, Felipe Segato, Matthew L. Freedman, Simon A. Gayther, Houtan Noushmehr, Kate Lawrenson. SOX18: A novel master regulator of high-grade serous ovarian tumorigenesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1494.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 2615-2615
    Abstract: Previous work indicates that identifying core transcription regulatory circuitries (CRCs) predicts tumor-specific genetic dependencies, in the form of transcription factors responsible for establishing this circuitry (Durbin, Zimmerman, Dharia, et al., 2018), which suggest targetable, onco-requisite factors. However, a systematic effort to identify CRCs across tumors has been challenging, since current approaches rely on data difficult to acquire in heterogeneous or low cell number samples. Here, we combine -omics approaches, including a newly developed algorithm to predict super-enhancer—driven CRCs in a range of tumor types. Our work leverages two characteristics CRC members: association with super-enhancers, defined using ChIP-Seq data, and highly cell type-specific expression, analyzed using the Cancer Cell-Type-Specificity (CaCTS) algorithm. CRC members uncovered with the CaCTS algorithm recover known drivers of specific tumor types and known misregulated pathways. We deeply characterized the CRC of high-grade serous ovarian cancer (HGSOC), a tumor type with poor overall prognosis for which few known driver mutations or targetable oncogenes are known. Our CRC models contain onco-requisite transcription factors that dominate the HGSOC gene expression program, including ESR1, MECOM (EVI1), and NOTCH2, each of which play important roles in other malignancies. Many of these CRC members represent HGSOC-selective dependencies in CRISPR/Cas9 screens. Profiling CRCs in HGSOC cells and presumed cells-of-origin (fallopian tube and ovarian tissue) highlights a set of CRC genes whose expression appears to have been evolved by HGSOC cells, including an isoform of the estrogen receptor. The dependence of HGSOC cells on transcription factors suggests a state of “transcriptional addiction,” which has been targeted with molecules against transcription apparatus. HGSOC lines are correspondingly susceptible to treatment with small molecules against transcriptional CDKs (CDK7 and CDK12). Also, some HGSOC-specific SEs are associated with genes druggable with existing small molecules. Thus, identifying the CRC governing HGSOC predicts dominant factors and suggests therapeutic avenues in this underserved disease. Similar predictions of CRCs in other poorly-studied tumors may also enhance understanding of those tumor cell identities and targetable nodes. Citation Format: Brian J. Abraham, Jessica Reddy, Marcos A. Fonseca, Isaac A. Klein, Lena K. Afeyan, Rosario I. Corona, Paloma Cejas, Felipe Segato, Beth Y. Karlan, Simon A. Gayther, Matthew L. Freedman, Houtan Noushmehr, Myles Brown, Ursula A. Matulonis, Kate Lawrenson, Richard A. Young. Systematic approaches to predict oncogenic transcriptional regulatory circuitries identify important nodes in high-grade serous ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2615.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
  • 8
    In: The American Journal of Human Genetics, Elsevier BV, ( 2024-5)
    Type of Medium: Online Resource
    ISSN: 0002-9297
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2024
    detail.hit.zdb_id: 1473813-2
    SSG: 12
    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...