8 results on '"Kate Lawrenson"'
Search Results
2. Epigenomic charting and functional annotation of risk loci in renal cell carcinoma
- Author
-
Amin H. Nassar, Sarah Abou Alaiwi, Sylvan C. Baca, Elio Adib, Rosario I. Corona, Ji-Heui Seo, Marcos A. S. Fonseca, Sandor Spisak, Talal El Zarif, Viktoria Tisza, David A. Braun, Heng Du, Monica He, Abdallah Flaifel, Michel Alchoueiry, Thomas Denize, Sayed G. Matar, Andres Acosta, Sachet Shukla, Yue Hou, John Steinharter, Gabrielle Bouchard, Jacob E. Berchuck, Edward O’Connor, Connor Bell, Pier Vitale Nuzzo, Gwo-Shu Mary Lee, Sabina Signoretti, Michelle S. Hirsch, Mark Pomerantz, Elizabeth Henske, Alexander Gusev, Kate Lawrenson, Toni K. Choueiri, David J. Kwiatkowski, and Matthew L. Freedman
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
While the mutational and transcriptional landscapes of renal cell carcinoma (RCC) are well-known, the epigenome is poorly understood. We characterize the epigenome of clear cell (ccRCC), papillary (pRCC), and chromophobe RCC (chRCC) by using ChIP-seq, ATAC-Seq, RNA-seq, and SNP arrays. We integrate 153 individual data sets from 42 patients and nominate 50 histology-specific master transcription factors (MTF) to define RCC histologic subtypes, including EPAS1 and ETS-1 in ccRCC, HNF1B in pRCC, and FOXI1 in chRCC. We confirm histology-specific MTFs via immunohistochemistry including a ccRCC-specific TF, BHLHE41. FOXI1 overexpression with knock-down of EPAS1 in the 786-O ccRCC cell line induces transcriptional upregulation of chRCC-specific genes, TFCP2L1, ATP6V0D2, KIT, and INSRR, implicating FOXI1 as a MTF for chRCC. Integrating RCC GWAS risk SNPs with H3K27ac ChIP-seq and ATAC-seq data reveals that risk-variants are significantly enriched in allelically-imbalanced peaks. This epigenomic atlas in primary human samples provides a resource for future investigation.
- Published
- 2023
- Full Text
- View/download PDF
3. Correction: Polygenic risk modeling for prediction of epithelial ovarian cancer risk
- Author
-
Daniel Barnes, Weiva Sieh, Jeffrey Weitzel, Paolo Peterlongo, Iain McNeish, Patricia Ganz, Allison Kurian, James Brenton, James Flanagan, Allison DePersia, Kate Lawrenson, Diether Lambrechts, Yin Ling Woo, Marc Goodman, Gareth Evans, Ana Peixoto, Katherine Nathanson, Mary Beth Terry, Jonathan Tyrer, Eileen Dareng, Zdeněk Kleibl, Cristina Rodriguez-Antona, Siel Olbrecht, Christoph Engel, and Niclas Håkansson
- Subjects
Genetics ,Genetics (clinical) - Published
- 2022
- Full Text
- View/download PDF
4. Reprogramming of the FOXA1 cistrome in treatment-emergent neuroendocrine prostate cancer
- Author
-
William C. Hahn, Xintao Qiu, Henry W. Long, Jacob E. Berchuck, Mark Pomerantz, Himisha Beltran, Klothilda Lim, Rand Arafeh, Matthew L. Freedman, Colm Morrissey, Sheng-Yu Ku, Bogdan Pasaniuc, Kathleen Kelly, Taylor E. Arnoff, Ilsa Coleman, Monica He, Lisha G. Brown, Holly M. Nguyen, Leigh Ellis, Amin Nassar, Eva Corey, Rosario I. Corona, Connor Bell, Sylvan C. Baca, Anjali V. Sheahan, Navonil De Sarkar, Kate Lawrenson, Claudia Giambartolomei, Keegan Korthauer, Arja Kaipainen, Myles Brown, Paloma Cejas, Peter S. Nelson, Supreet Agarwal, David Y. Takeda, Amina Zoubeidi, Sarah Abou Alaiwi, Marcos A. S. Fonseca, Edward P. O’Connor, Justin H. Hwang, and Ji-Heui Seo
- Subjects
Male ,Epigenomics ,0301 basic medicine ,medicine.medical_treatment ,General Physics and Astronomy ,Androgen ,Targeted therapy ,Prostate cancer ,0302 clinical medicine ,Receptors ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Tumor ,Multidisciplinary ,Prostate Cancer ,Chromatin binding ,Gene Expression Regulation, Neoplastic ,Neuroendocrine Tumors ,Cistrome ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Disease Progression ,RNA Interference ,Hepatocyte Nuclear Factor 3-alpha ,Urologic Diseases ,Science ,Adenocarcinoma ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Transcription factor ,Neoplastic ,Human Genome ,Prostatic Neoplasms ,General Chemistry ,medicine.disease ,Androgen receptor ,030104 developmental biology ,Gene Expression Regulation ,Mutation ,Cancer research ,FOXA1 ,Chromatin immunoprecipitation - Abstract
Lineage plasticity, the ability of a cell to alter its identity, is an increasingly common mechanism of adaptive resistance to targeted therapy in cancer. An archetypal example is the development of neuroendocrine prostate cancer (NEPC) after treatment of prostate adenocarcinoma (PRAD) with inhibitors of androgen signaling. NEPC is an aggressive variant of prostate cancer that aberrantly expresses genes characteristic of neuroendocrine (NE) tissues and no longer depends on androgens. Here, we investigate the epigenomic basis of this resistance mechanism by profiling histone modifications in NEPC and PRAD patient-derived xenografts (PDXs) using chromatin immunoprecipitation and sequencing (ChIP-seq). We identify a vast network of cis-regulatory elements (N~15,000) that are recurrently activated in NEPC. The FOXA1 transcription factor (TF), which pioneers androgen receptor (AR) chromatin binding in the prostate epithelium, is reprogrammed to NE-specific regulatory elements in NEPC. Despite loss of dependence upon AR, NEPC maintains FOXA1 expression and requires FOXA1 for proliferation and expression of NE lineage-defining genes. Ectopic expression of the NE lineage TFs ASCL1 and NKX2-1 in PRAD cells reprograms FOXA1 to bind to NE regulatory elements and induces enhancer activity as evidenced by histone modifications at these sites. Our data establish the importance of FOXA1 in NEPC and provide a principled approach to identifying cancer dependencies through epigenomic profiling., The molecular processes that lead to neuroendocrine prostate cancer after treating prostate adenocarcinoma (PRAD) are not well understood. Here the authors show that regulation by FOXA1 and changes in the epigenomic profile drive the transition from PRAD to a neuroendocrine phenotype.
- Published
- 2021
- Full Text
- View/download PDF
5. CAUSEL: an epigenome- and genome-editing pipeline for establishing function of noncoding GWAS variants
- Author
-
Christopher A. Haiman, J. Keith Joung, Kate Lawrenson, Ying Han, Zachery T Herbert, Zoltan Szallasi, Rebecca Tayler Cottman, Qiyuan Li, Mark Pomerantz, Viktoria Tisza, István Csabai, Simon A. Gayther, Norbert Solymosi, Ji-Heui Seo, Romina E. Lenci, Matthew L. Freedman, Yanfang Fu, Sándor Spisák, and Matthew S. Chabot
- Subjects
Epigenomics ,Male ,Regulatory Factor X Transcription Factors ,Locus (genetics) ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Genome ,Article ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Genome editing ,Cell Line, Tumor ,Epigenome editing ,Humans ,Genetic Predisposition to Disease ,RNA, Messenger ,Alleles ,Homeodomain Proteins ,Genetics ,Transcription activator-like effector nuclease ,Reverse Transcriptase Polymerase Chain Reaction ,Chromosome Mapping ,Prostatic Neoplasms ,General Medicine ,Epigenome ,3. Good health ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Histone Code ,Chromosomes, Human, Pair 6 ,Genome-Wide Association Study ,Transcription Factors - Abstract
The vast majority of disease-associated single-nucleotide polymorphisms (SNPs) mapped by genome-wide association studies (GWASs) are located in the non-protein-coding genome, but establishing the functional and mechanistic roles of these sequence variants has proven challenging. Here we describe a general pipeline in which candidate functional SNPs are first evaluated by fine mapping, epigenomic profiling, and epigenome editing, and then interrogated for causal function by using genome editing to create isogenic cell lines followed by phenotypic characterization. To validate this approach, we analyzed the 6q22.1 prostate cancer risk locus and identified rs339331 as the top-scoring SNP. Epigenome editing confirmed that the rs339331 region possessed regulatory potential. By using transcription activator-like effector nuclease (TALEN)-mediated genome editing, we created a panel of isogenic 22Rv1 prostate cancer cell lines representing all three genotypes (TT, TC, CC) at rs339331. Introduction of the 'T' risk allele increased transcription of the regulatory factor 6 (RFX6) gene, increased homeobox B13 (HOXB13) binding at the rs339331 region, and increased deposition of the enhancer-associated H3K4me2 histone mark at the rs339331 region compared to lines homozygous for the 'C' protective allele. The cell lines also differed in cellular morphology and adhesion, and pathway analysis of differentially expressed genes suggested an influence of androgens. In summary, we have developed and validated a widely accessible approach that can be used to establish functional causality for noncoding sequence variants identified by GWASs.
- Published
- 2015
- Full Text
- View/download PDF
6. Genome-wide significant risk associations for mucinous ovarian carcinoma
- Author
-
Jenny Lester, Arif B. Ekici, Xiao-Ou Shu, Georgia Chenevix-Trench, Julie M. Cunningham, Catherine M. Phelan, Tassja J. Spindler, Shelley S. Tworoger, Allan Jensen, Diana Eccles, Wei Zheng, Satoyo Hosono, Svend Aage Engelholm, Linda S. Cook, Thomas A. Sellers, John R. McLaughlin, Matthias W. Beckmann, Elizabeth M. Poole, Ignace Vergote, Francesmary Modugno, Jennifer A. Doherty, Sandrina Lambrechts, Lara Sucheston, Wang Gohrke Shan, Rosalind Glasspool, Barry P. Rosen, Pamela J. Thompson, Christine Walsh, Camilla Krakstad, Jennifer Permuth-Wey, Hoda Anton-Culver, Ralf Bützow, Ed Dicks, Yin Ling Woo, Honglin Song, Qiyuan Li, Leon F.A.G. Massuger, Yurii B. Shvetsov, Joellen M. Schildkraut, Xiaoqing Chen, Hannah P. Yang, Graham G. Giles, Sara H. Olson, Edwin S. Iversen, Claus Høgdall, Jacek Grownwald, Fiona Bruinsma, Peter Hillemanns, Iwona K. Rzepecka, Douglas T. Easton, Robert P. Edwards, Lynne R. Wilkens, Yukie Bean, Y. Ann Chen, Jonathan Tyrer, Tanja Pejovic, Patricia Harrington, Iain A. McNeish, Steven A. Narod, Joanna Moes-Sosnowska, Hanis Nazihah Hasmad, Ji-Heui Seo, Line Bjørge, Peter A. Fasching, Daniel W. Cramer, Florian Heitz, Beth Y. Karlan, Cezary Cybulski, Simon A. Gayther, Jolanta Kupryjanczyk, Nhu D. Le, Alexander Hein, Soo Hwang Teo, Nicolas Wentzensen, Valerie McGuire, Michelle A.T. Hildebrandt, Argyrios Ziogas, Heli Nevanlinna, Karen H. Lu, Diether Lambrechts, Kirsten B. Moysich, Jonathan Beesley, Kate Lawrenson, Ian G. Campbell, Andrew Berchuck, Philipp Harter, Lene Lundvall, Sandra Orsulic, Kathryn L. Terry, Jolanta Lissowska, Helen Baker, Matthew L. Freedman, Linda E. Kelemen, Maria Bisogna, Celeste Leigh Pearce, Ira Schwaab, Elisa V. Bandera, Kristine G. Wicklund, Arto Leminen, Nadeem Siddiqui, Joseph H. Rothstein, Melissa Kellar, Matthias Dürst, Harvey A. Risch, Włodzimierz Sawicki, Katja K.H. Aben, Alice S. Whittemore, Kunle Odunsi, Shashi Lele, Noor Azmi Mat Adenan, Susanne K. Kjaer, Robert A. Vierkant, Jan Lubinski, Natalia Bogdanova, Helga B. Salvesen, Anja Rudolph, Els Van Nieuwenhuysen, Irene Orlow, Joe Dennis, Ingvild L. Tangen, Estrid Høgdall, Alice W. Lee, Roberta B. Ness, James Paul, Malcolm C. Pike, Rachel Palmieri Weber, Anna H. Wu, Agnieszka Dansonka-Mieszkowska, Melissa C. Southey, Natalia Antonenkova, Anne M. van Altena, Xifeng Wu, Keitaro Matsuo, Anna Jakubowska, Paul D.P. Pharoah, Weiva Sieh, Lambertus A. Kiemeney, Lotte Nedergaard, Ursula Eilber, Janet M. Lee, Zhihua Chen, Andreas du Bois, Joseph L. Kelley, Aleksandra Gentry-Maharaj, Jenny Chang-Claude, Ellen L. Goode, Bu Tian Ji, Liisa M. Pelttari, Usha Menon, Dong Liang, Yu-Tang Gao, Brooke L. Fridley, Mary Anne Rossing, Thilo Dörk, Douglas A. Levine, Louise A. Brinton, Marc T. Goodman, Angela Brooks-Wilson, Ingo B. Runnebaum, Karen Carty, and Susan J. Ramus
- Subjects
Green Fluorescent Proteins ,Molecular Sequence Data ,Quantitative Trait Loci ,Single-nucleotide polymorphism ,Genome-wide association study ,Quantitative trait locus ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Cell Line, Tumor ,Ovarian carcinoma ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Association Studies ,030304 developmental biology ,Genetic association ,Cell Nucleus ,Homeodomain Proteins ,Ovarian Neoplasms ,0303 health sciences ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,medicine.disease ,Adenocarcinoma, Mucinous ,Neoplasm Proteins ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,3. Good health ,Gene Expression Regulation, Neoplastic ,Microscopy, Fluorescence ,Chromosomes, Human, Pair 2 ,Urological cancers Radboud Institute for Health Sciences [Radboudumc 15] ,030220 oncology & carcinogenesis ,Expression quantitative trait loci ,Cancer research ,Adenocarcinoma ,Female ,Ovarian cancer ,Genome-Wide Association Study - Abstract
Genome-wide association studies have identified several risk associations for ovarian carcinomas (OC) but not for mucinous ovarian carcinomas (MOC). Genotypes from OC cases and controls were imputed into the 1000 Genomes Project reference panel. Analysis of 1,644 MOC cases and 21,693 controls identified three novel risk associations: rs752590 at 2q13 (P = 3.3 × 10−8), rs711830 at 2q31.1 (P = 7.5 × 10−12) and rs688187 at 19q13.2 (P = 6.8 × 10−13). Expression Quantitative Trait Locus (eQTL) analysis in ovarian and colorectal tumors (which are histologically similar to MOC) identified significant eQTL associations for HOXD9 at 2q31.1 in ovarian (P = 4.95 × 10−4, FDR = 0.003) and colorectal (P = 0.01, FDR = 0.09) tumors, and for PAX8 at 2q13 in colorectal tumors (P = 0.03, FDR = 0.09). Chromosome conformation capture analysis identified interactions between the HOXD9 promoter and risk SNPs at 2q31.1. Overexpressing HOXD9 in MOC cells augmented the neoplastic phenotype. These findings provide the first evidence for MOC susceptibility variants and insights into the underlying biology of the disease.
- Published
- 2015
- Full Text
- View/download PDF
7. Clinical Implications of Marker Expression of Carcinoma-Associated Fibroblasts (CAFs) in Patients with Epithelial Ovarian Carcinoma After Treatment with Neoadjuvant Chemotherapy
- Author
-
Tanja Pejovic, Kate Lawrenson, Teodulo Meneses, Annie A. Yessaian, Damanzoopinder Samrao, Paulette Mhawech-Fauceglia, Dan Wang, Grace Kim, and Song Liu
- Subjects
Oncology ,Original Paper ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,medicine.medical_treatment ,Tumor initiation ,Malignancy ,medicine.disease ,Debulking ,digestive system diseases ,Targeted therapy ,Exact test ,Stroma ,Internal medicine ,Medicine ,Immunohistochemistry ,business ,Neoadjuvant therapy - Abstract
Cancer-associated fibroblasts (CAFs) play an important role in tumor initiation and progression. The aim of this study is to explore the role of 2 CAF markers, fibroblast activated protein (FAP) and α-smooth muscle actin (αSMA), in patients with epithelial ovarian cancer (EOC) post-neoadjuvant chemotherapy. Sixty-six patients with the diagnosis of EOC treated with debulking surgery after neoadjuvant therapy were retrieved from the archives. Immunohistochemistry for FAP and αSMA antibodies were performed on paraffin-embedded tissue. Fisher's exact test was performed to test the association between FAP and αSMA expression and disease status. Kaplan-Meier method with log-rank test was used to check the survival difference between different FAP tumor/stroma expressions. FAP stroma (pos) . expression was strongly associated with higher recurrences rate [OR: 15.95; 95 % CI: 1.521-835.206; p = 0.0072]. Cases with combined FAP stroma (pos) and FAP tumor (neg) had higher death rate [OR: 4.845; 95 % CI: 1.53-16.61; p = 0.0046] and higher recurrence rate [OR: 5.12; 95 % CI: 0.91-54.42; p = 0.0487] compared to all the others. Cases with combined FAP stroma (neg) and FAP tumor (neg) were more likely to have lower recurrence rates [OR: 0.086; 95 % CI: 0.001-0.997; p = 0.0248]. αSMA was expressed by tumor-associated stroma in 95 % of cases and by tumor cells in 9 % of cases. No statistical power was found for αSMA and disease status. Our data indicate that FAP plays an important role in predicting tumor aggressiveness in patients with EOC post-neoadjuvant therapy, and its frequent expression in this malignancy implicates that FAP targeted therapy could be a very attractive strategy.
- Published
- 2013
- Full Text
- View/download PDF
8. Targeting Src in endometriosis-associated ovarian cancer
- Author
-
Heidi M. Sowter, Elham Pakzamir, Paulette Mhawech-Fauceglia, Roxanne Manek, Simon A. Gayther, Tanja Pejovic, and Kate Lawrenson
- Subjects
0301 basic medicine ,Cancer Research ,Cancer ,Biology ,Cell cycle ,medicine.disease ,medicine.disease_cause ,Molecular oncology ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Growth factor receptor ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Original Article ,Src family kinase ,Ovarian cancer ,Carcinogenesis ,Molecular Biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The SRC proto-oncogene is commonly overexpressed or activated during cancer development. Src family kinase inhibitors are approved for the treatment of certain leukemias, and are in clinical trials for the treatment of solid tumors. Src signaling is activated in endometriosis, a precursor of clear cell and endometrioid subtypes of epithelial ovarian cancers (OCs). We examined the expression of phosphorylated Src (Src-pY416) in 381 primary OC tissues. Thirty-six percent of OCs expressed Src-pY416. Src-pY416 expression was most common in endometriosis-associated OCs (EAOCs) (P=0.011), particularly in clear cell OCs where 58.5% of cases expressed Src-pY416. Src-pY416 expression was associated with shorter overall survival (log rank P=0.002). In vitro inhibition of Src signaling using 4-amino-5-(4-chlorophenyl)-7-(dimethylethyl)pyrazolo[3,4-d]pyrimidine (PP2) resulted in reduced anchorage-independent and -dependent growth, and in three-dimensional cell culture models PP2 disrupted aggregate formation in Src-pY416-positive but not in Src-pY416-negative cell lines. These data suggest that targeting active Src signaling could be a novel therapeutic opportunity for EAOCs, and support the further pre-clinical investigation of Src family kinase inhibitors for treating OCs expressing Src-pY416.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.