25 results on '"Marcin Cieślik"'
Search Results
2. Data from Cancer Cell Intrinsic and Immunologic Phenotypes Determine Clinical Outcomes in Basal-like Breast Cancer
- Author
-
Arul M. Chinnaiyan, Dan R. Robinson, Jamie Guenthoer, Peggy Porter, Xuhong Cao, Mei-Tzu C. Tang, Erin Cobain, Lanbo Xiao, Yi-Mi Wu, Marcin Cieślik, Yuping Zhang, and Christopher I. Li
- Abstract
Purpose:Basal-like breast cancer (BLBC) is a particularly aggressive intrinsic molecular subtype of breast cancer that lacks targeted therapies. There is also no clinically useful test to risk stratify patients with BLBC. We hypothesized that a transcriptome-based phenotypic characterization of BLBC tumors and their microenvironments may overcome these challenges.Experimental Design:We conducted a retrospective correlative genomic sequencing study using a matched pairs design with validation in five independent cohorts. The study was conducted on a large population-based prospective cohort of the major molecular subtypes of breast cancer conducted in the greater Seattle-Puget Sound metropolitan area. Cases consisted of women 20–69 years of age first diagnosed with invasive breast cancer identified through the population-based Surveillance Epidemiology and End Results program. Patients for this analysis (n = 949) were identified from the 1,408 patients with stage I–III triple-negative breast cancer [estrogen receptor–negative (ER−), progesterone receptor–negative (PR−), HER2−]. Of the 949 women, 248 developed a recurrence after their initial diagnosis. A matched set of 67 recurrent and nonrecurrent BLBC tumors was subjected to transcriptome sequencing. Through RNA sequencing of the matched sets of recurrent and nonrecurrent BLBC tumors, we aimed to identify prognostic phenotypes.To identify nonredundant and uncorrelated prognostic genes, we used an ensemble of variable selection algorithms, which resulted in a ranking of genes on the basis of their expected utility in classification. Using leave-one-out cross-validation, we trained a random forest classifier on the basis of the top 21 genes (BRAVO-DX). Validations were performed in five independent triple-negative or BLBC cohorts, and biomarker robustness and transferability were demonstrated by employing real-time PCR.Results:We found that cancer cell intrinsic and immunologic phenotypes are independent predictors of recurrence. By simultaneously interrogating the tumor and its microenvironment, we developed a compound risk model that stratified patients into low-, medium-, and high-risk groups, with a 14%/56%/74% chance of recurrence, respectively. Biologically, the primary tumors of patients who developed a recurrence had increased growth factor signaling and stem-like features, while nonrecurrent tumors showed high lymphocyte infiltration with clonal expansion of T and B cells, as well as antitumor polarization of macrophages. We validated our model in five independent cohorts, including three large cohorts, where BRAVO-DX was highly informative in identifying patients with disease recurrence [HR, 6.79 (95% confidence interval (CI), 1.89–24.37); HR, 3.45 (95% CI, 2.41–4.93); and HR, 1.69 (95% CI, 1.17–2.46)]. A smaller gene set focused on the tumor immunophenotype, BRAVO-IMMUNE, was highly prognostic in all five cohorts.Conclusions:Together, these results indicate that phenotypic characteristics of BLBCs and their microenvironment are associated with recurrence-free survival and demonstrate the utility of intrinsic and extrinsic phenotypes as independent prognostic biomarkers in BLBC. Pending further evaluation and validation, our prognostic model has the potential to inform clinical decision-making for patients with BLBC as it identifies those at high risk of rapidly progressing on standard chemotherapy, as well as those who may benefit from alternative first-line therapies.
- Published
- 2023
3. Supplementary Figures S1-S10 from Cancer Cell Intrinsic and Immunologic Phenotypes Determine Clinical Outcomes in Basal-like Breast Cancer
- Author
-
Arul M. Chinnaiyan, Dan R. Robinson, Jamie Guenthoer, Peggy Porter, Xuhong Cao, Mei-Tzu C. Tang, Erin Cobain, Lanbo Xiao, Yi-Mi Wu, Marcin Cieślik, Yuping Zhang, and Christopher I. Li
- Abstract
Supplementary Figures S1 through S10
- Published
- 2023
4. Cancer Cell Intrinsic and Immunologic Phenotypes Determine Clinical Outcomes in Basal-like Breast Cancer
- Author
-
Lanbo Xiao, Yuping Zhang, Arul M. Chinnaiyan, Christopher I. Li, Dan R. Robinson, Marcin Cieślik, Yi-Mi Wu, Jamie Guenthoer, Xuhong Cao, Peggy L. Porter, Mei-Tzu C. Tang, and Erin F. Cobain
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,Disease ,medicine.disease ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Internal medicine ,Surveillance, Epidemiology, and End Results ,Medicine ,Biomarker (medicine) ,Stage (cooking) ,business ,Prospective cohort study ,education - Abstract
Purpose: Basal-like breast cancer (BLBC) is a particularly aggressive intrinsic molecular subtype of breast cancer that lacks targeted therapies. There is also no clinically useful test to risk stratify patients with BLBC. We hypothesized that a transcriptome-based phenotypic characterization of BLBC tumors and their microenvironments may overcome these challenges. Experimental Design: We conducted a retrospective correlative genomic sequencing study using a matched pairs design with validation in five independent cohorts. The study was conducted on a large population-based prospective cohort of the major molecular subtypes of breast cancer conducted in the greater Seattle-Puget Sound metropolitan area. Cases consisted of women 20–69 years of age first diagnosed with invasive breast cancer identified through the population-based Surveillance Epidemiology and End Results program. Patients for this analysis (n = 949) were identified from the 1,408 patients with stage I–III triple-negative breast cancer [estrogen receptor–negative (ER−), progesterone receptor–negative (PR−), HER2−]. Of the 949 women, 248 developed a recurrence after their initial diagnosis. A matched set of 67 recurrent and nonrecurrent BLBC tumors was subjected to transcriptome sequencing. Through RNA sequencing of the matched sets of recurrent and nonrecurrent BLBC tumors, we aimed to identify prognostic phenotypes. To identify nonredundant and uncorrelated prognostic genes, we used an ensemble of variable selection algorithms, which resulted in a ranking of genes on the basis of their expected utility in classification. Using leave-one-out cross-validation, we trained a random forest classifier on the basis of the top 21 genes (BRAVO-DX). Validations were performed in five independent triple-negative or BLBC cohorts, and biomarker robustness and transferability were demonstrated by employing real-time PCR. Results: We found that cancer cell intrinsic and immunologic phenotypes are independent predictors of recurrence. By simultaneously interrogating the tumor and its microenvironment, we developed a compound risk model that stratified patients into low-, medium-, and high-risk groups, with a 14%/56%/74% chance of recurrence, respectively. Biologically, the primary tumors of patients who developed a recurrence had increased growth factor signaling and stem-like features, while nonrecurrent tumors showed high lymphocyte infiltration with clonal expansion of T and B cells, as well as antitumor polarization of macrophages. We validated our model in five independent cohorts, including three large cohorts, where BRAVO-DX was highly informative in identifying patients with disease recurrence [HR, 6.79 (95% confidence interval (CI), 1.89–24.37); HR, 3.45 (95% CI, 2.41–4.93); and HR, 1.69 (95% CI, 1.17–2.46)]. A smaller gene set focused on the tumor immunophenotype, BRAVO-IMMUNE, was highly prognostic in all five cohorts. Conclusions: Together, these results indicate that phenotypic characteristics of BLBCs and their microenvironment are associated with recurrence-free survival and demonstrate the utility of intrinsic and extrinsic phenotypes as independent prognostic biomarkers in BLBC. Pending further evaluation and validation, our prognostic model has the potential to inform clinical decision-making for patients with BLBC as it identifies those at high risk of rapidly progressing on standard chemotherapy, as well as those who may benefit from alternative first-line therapies.
- Published
- 2021
5. A novel ATXN1-DUX4 fusion expands the spectrum of ‘CIC-rearranged sarcoma’ of the CNS to include non-CIC alterations
- Author
-
Rohit Mehra, Martha Quezado, Rajen Mody, Kenneth Aldape, Marcin Cieślik, Drew Pratt, Andrea Franson, Arul M. Chinnaiyan, Evan Cantor, Hong Xiao, Zied Abdullaev, Chandan Kumar-Sinha, Lina Shao, and Sandra Camelo-Piragua
- Subjects
Homeodomain Proteins ,Male ,CIC-Rearranged Sarcoma ,Oncogene Proteins, Fusion ,Brain Neoplasms ,business.industry ,Sarcoma ,Biology ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Text mining ,DUX4 ,Child, Preschool ,Cancer research ,Humans ,Neurology (clinical) ,business ,Ataxin-1 - Published
- 2021
6. Cancer Cell Intrinsic and Immunologic Phenotypes Determine Clinical Outcomes in Basal-like Breast Cancer
- Author
-
Christopher I, Li, Yuping, Zhang, Marcin, Cieślik, Yi-Mi, Wu, Lanbo, Xiao, Erin, Cobain, Mei-Tzu C, Tang, Xuhong, Cao, Peggy, Porter, Jamie, Guenthoer, Dan R, Robinson, and Arul M, Chinnaiyan
- Subjects
Adult ,Washington ,Whole Genome Sequencing ,Breast Neoplasms ,Middle Aged ,Pregnanes ,Prognosis ,Phenotype ,Tumor Microenvironment ,Humans ,Female ,Glycosides ,Prospective Studies ,Neoplasm Recurrence, Local ,Transcriptome ,Aged - Abstract
Basal-like breast cancer (BLBC) is a particularly aggressive intrinsic molecular subtype of breast cancer that lacks targeted therapies. There is also no clinically useful test to risk stratify patients with BLBC. We hypothesized that a transcriptome-based phenotypic characterization of BLBC tumors and their microenvironments may overcome these challenges.We conducted a retrospective correlative genomic sequencing study using a matched pairs design with validation in five independent cohorts. The study was conducted on a large population-based prospective cohort of the major molecular subtypes of breast cancer conducted in the greater Seattle-Puget Sound metropolitan area. Cases consisted of women 20-69 years of age first diagnosed with invasive breast cancer identified through the population-based Surveillance Epidemiology and End Results program. Patients for this analysis (We found that cancer cell intrinsic and immunologic phenotypes are independent predictors of recurrence. By simultaneously interrogating the tumor and its microenvironment, we developed a compound risk model that stratified patients into low-, medium-, and high-risk groups, with a 14%/56%/74% chance of recurrence, respectively. Biologically, the primary tumors of patients who developed a recurrence had increased growth factor signaling and stem-like features, while nonrecurrent tumors showed high lymphocyte infiltration with clonal expansion of T and B cells, as well as antitumor polarization of macrophages. We validated our model in five independent cohorts, including three large cohorts, where BRAVO-DX was highly informative in identifying patients with disease recurrence [HR, 6.79 (95% confidence interval (CI), 1.89-24.37); HR, 3.45 (95% CI, 2.41-4.93); and HR, 1.69 (95% CI, 1.17-2.46)]. A smaller gene set focused on the tumor immunophenotype, BRAVO-IMMUNE, was highly prognostic in all five cohorts.Together, these results indicate that phenotypic characteristics of BLBCs and their microenvironment are associated with recurrence-free survival and demonstrate the utility of intrinsic and extrinsic phenotypes as independent prognostic biomarkers in BLBC. Pending further evaluation and validation, our prognostic model has the potential to inform clinical decision-making for patients with BLBC as it identifies those at high risk of rapidly progressing on standard chemotherapy, as well as those who may benefit from alternative first-line therapies.
- Published
- 2020
7. Analysis of the androgen receptor–regulated lncRNA landscape identifies a role for ARLNC1 in prostate cancer progression
- Author
-
Susan M. Freier, Mats Ljungman, Lanbo Xiao, Sahr Yazdani, Kristin M. Juckette, Andrew T. Watt, Mona Batish, Shuling Guo, Alexander R. Gawronski, Saravana M. Dhanasekaran, Sudhanshu Shukla, John T. Wei, Michael Uhl, Rohit Malik, Hui Jiang, Yasuyuki Hosono, Yashar S. Niknafs, Yuanyuan Qiao, Utsav Patel, Sethuramasundaram Pitchiaya, Rohit Mehra, Lakshmi P. Kunju, Michelle T. Paulsen, Felix Y. Feng, Rolf Backofen, Xia Jiang, Xuhong Cao, Shruthi Subramaniam, Cenk Sahinalp, Tzu-Ying Liu, Jean C.-Y. Tien, Matthew K. Iyer, Girish C. Shukla, Arul M. Chinnaiyan, Yajia Zhang, Marcin Cieślik, and Lisha Wang
- Subjects
Male ,0301 basic medicine ,Biology ,Medical and Health Sciences ,Article ,Cell Line ,Androgen ,Transcriptome ,03 medical and health sciences ,Prostate cancer ,Prostate ,androgen receptor ,Cell Line, Tumor ,Receptors ,Genetics ,medicine ,Humans ,Regulation of gene expression ,Neoplastic ,Gene knockdown ,Tumor ,long non-coding RNA ,ARLNC1 ,Prostatic Neoplasms ,Biological Sciences ,prostate cancer ,medicine.disease ,Long non-coding RNA ,3. Good health ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Receptors, Androgen ,Disease Progression ,Androgens ,Cancer research ,RNA ,RNA, Long Noncoding ,Long Noncoding ,Signal transduction ,Signal Transduction ,Developmental Biology - Abstract
The androgen receptor (AR) plays a critical role in the development of the normal prostate as well as prostate cancer. Using an integrative transcriptomic analysis of prostate cancer cell lines and tissues, we identified ARLNC1 (AR-regulated long noncoding RNA 1) as an important long noncoding RNA that is strongly associated with AR signaling in prostate cancer progression. Not only was ARLNC1 induced by the AR protein, but ARLNC1 stabilized the AR transcript via RNA-RNA interaction. ARLNC1 knockdown suppressed AR expression, global AR signaling and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARLNC1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression and identify ARLNC1 as a novel therapeutic target.
- Published
- 2018
8. Cancer transcriptome profiling at the juncture of clinical translation
- Author
-
Marcin Cieślik and Arul M. Chinnaiyan
- Subjects
0301 basic medicine ,Sequence Analysis, RNA ,Sequence analysis ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,Cancer ,Context (language use) ,Translation (biology) ,Computational biology ,Biology ,medicine.disease ,Phenotype ,Neoplasm genetics ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,Neoplasms ,Genetics ,medicine ,Animals ,Humans ,Transcriptome profiling ,RNA, Neoplasm ,Molecular Biology ,Genetics (clinical) - Abstract
Methodological breakthroughs over the past four decades have repeatedly revolutionized transcriptome profiling. Using RNA sequencing (RNA-seq), it has now become possible to sequence and quantify the transcriptional outputs of individual cells or thousands of samples. These transcriptomes provide a link between cellular phenotypes and their molecular underpinnings, such as mutations. In the context of cancer, this link represents an opportunity to dissect the complexity and heterogeneity of tumours and to discover new biomarkers or therapeutic strategies. Here, we review the rationale, methodology and translational impact of transcriptome profiling in cancer.
- Published
- 2017
9. Development of Peptidomimetic Inhibitors of the ERG Gene Fusion Product in Prostate Cancer
- Author
-
Bushra Ateeq, Irfan A. Asangani, Wei Yan, Jean Ching Yi Tien, Xiaoju Wang, Shaomeng Wang, Ingrid J. Apel, Sooryanarayana Varambally, Cynthia Wang, Balabhadrapatruni V. S. K. Chakravarthi, Anton Poliakov, Yuanyuan Qiao, Arul M. Chinnaiyan, Kristin M. Juckette, Sethuramasundaram Pitchiaya, Rui Wang, Hui Jiang, Marcin Cieślik, Xuhong Cao, and Xiaojun Jing
- Subjects
Male ,0301 basic medicine ,Cancer Research ,genetic structures ,Oncogene Proteins, Fusion ,Peptidomimetic ,Mice, Nude ,Neovascularization, Physiologic ,Antineoplastic Agents ,Chick Embryo ,Biology ,Pharmacology ,Bioinformatics ,medicine.disease_cause ,Fusion gene ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Protein Domains ,Transcriptional Regulator ERG ,Transcription (biology) ,Peptide Library ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Transcription factor ,business.industry ,Prostatic Neoplasms ,DNA-binding domain ,DNA ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,eye diseases ,Chromatin ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Product (mathematics) ,Cancer cell ,Cancer research ,sense organs ,Peptidomimetics ,Carcinogenesis ,business ,Erg - Abstract
Transcription factors play a key role in the development of diverse cancers, and therapeutically targeting them has remained a challenge. In prostate cancer, the gene encoding the transcription factor ERG is recurrently rearranged and plays a critical role in prostate oncogenesis. Here, we identified a series of peptides that interact specifically with the DNA binding domain of ERG. ERG inhibitory peptides (EIPs) and derived peptidomimetics bound ERG with high affinity and specificity, leading to proteolytic degradation of the ERG protein. The EIPs attenuated ERG-mediated transcription, chromatin recruitment, protein-protein interactions, cell invasion and proliferation, and tumor growth. Thus, peptidomimetic targeting of transcription factor fusion products may provide a promising therapeutic strategy for prostate cancer as well as other malignancies.
- Published
- 2017
10. Epigenetic driver mutations in ARID1A shape cancer immune phenotype and immunotherapy
- Author
-
Michael D. Green, Arul M. Chinnaiyan, Weiping Zou, Lili Zhao, Weimin Wang, Kathleen R. Cho, Mengyao Tan, Jipeng Guo, Heng Lin, Weichao Wang, Jiajia Zhou, David G. Huntsman, Yali Dou, Shuang Wei, Yajia Zhang, Xiaojun Jing, Marcin Cieślik, Timothy A. Chan, Linda Vatan, Gaopeng Li, Rugang Zhang, Jing Li, Wei Li, Ilona Kryczek, and Benjamin G. Bitler
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Biology ,Chromatin remodeling ,Epigenesis, Genetic ,Immunophenotyping ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Epigenetics ,Melanoma ,Ovarian Neoplasms ,EZH2 ,General Medicine ,Immunotherapy ,Chromatin Assembly and Disassembly ,Chromatin ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Tumor Escape ,Interferons ,Signal Transduction ,Transcription Factors ,Research Article - Abstract
Whether mutations in cancer driver genes directly affect cancer immune phenotype and T cell immunity remains a standing question. ARID1A is a core member of the polymorphic BRG/BRM-associated factor chromatin remodeling complex. ARID1A mutations occur in human cancers and drive cancer development. Here, we studied the molecular, cellular, and clinical impact of ARID1A aberrations on cancer immunity. We demonstrated that ARID1A aberrations resulted in limited chromatin accessibility to IFN-responsive genes, impaired IFN gene expression, anemic T cell tumor infiltration, poor tumor immunity, and shortened host survival in many human cancer histologies and in murine cancer models. Impaired IFN signaling was associated with poor immunotherapy response. Mechanistically, ARID1A interacted with EZH2 via its carboxyl terminal and antagonized EZH2-mediated IFN responsiveness. Thus, the interaction between ARID1A and EZH2 defines cancer IFN responsiveness and immune evasion. Our work indicates that cancer epigenetic driver mutations can shape cancer immune phenotype and immunotherapy.
- Published
- 2019
11. Next-generation RNA Sequencing-based Biomarker Characterization of Chromophobe Renal Cell Carcinoma and Related Oncocytic Neoplasms
- Author
-
Lisha Wang, Pankaj Vats, Saravana M. Dhanasekaran, Fengyun Su, Jin Chen, Pedram Argani, Thomas J. Giordano, Rahul Mannan, Arul M. Chinnaiyan, Xiaoming Wang, Sathiya Pandi Narayanan, Rohit Mehra, Stephanie L. Skala, Xuhong Cao, Marcin Cieślik, Yuping Zhang, and Javed Siddiqui
- Subjects
Pathology ,medicine.medical_specialty ,Urology ,Chromophobe Renal Cell Carcinoma ,030232 urology & nephrology ,Chromophobe cell ,Gene mutation ,urologic and male genital diseases ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,medicine ,Humans ,Oncocytoma ,Carcinoma, Renal Cell ,biology ,business.industry ,Sequence Analysis, RNA ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Kidney Neoplasms ,RHCG ,030220 oncology & carcinogenesis ,biology.protein ,Biomarker (medicine) ,Immunohistochemistry ,business - Abstract
Background Renal cell carcinomas (RCCs) are a heterogeneous group of neoplasms. Recent sequencing studies revealed various molecular features associated with histologic RCC subtypes, including chromophobe renal cell carcinoma (ChRCC). Objective To characterize the gene expression and biomarker signatures associated with ChRCC. Design, setting, and participants We performed integrative analysis on RNA sequencing data available from 1049 RCC specimens from The Cancer Genome Atlas and in-house studies. Our workflow identified genes relatively enriched in ChRCC, including Forkhead box I1 (FOXI1), Rh family C glycoprotein (RHCG), and LINC01187. We assessed the expression pattern of FOXI1 and RHCG protein by immunohistochemistry (IHC) and LINC01187 mRNA by RNA in situ hybridization (RNA-ISH) in whole tissue sections representing a cohort of 197 RCC cases, including both primary and metastatic tumors. Outcome measurements and statistical analysis The FOXI1 and RHCG IHC staining, as well as the LINC01187 RNA-ISH staining, was evaluated in each case for intensity, pattern, and localization of expression. Results and limitations All primary and metastatic classic ChRCCs demonstrated homogeneous positive labeling for FOXI1, RHCG proteins, and LINC01187 transcript. Unclassified RCC with oncocytic features, oncocytoma, and hybrid oncocytic tumor, as well as all but two cases of eosinophilic ChRCC also stained positive. Importantly, metastatic and primary RCC of all other subtypes did not demonstrate any unequivocal staining for FOXI1, RHCG, or LINC01187. In normal kidney, FOXI1, RHCG, and LINC01187 were detected in the distal nephron segment, specifically in intercalated cells. Two cases of eosinophilic ChRCC with focal expression of FOXI1 and LINC01187, and Golgi-like RHCG staining were found to contain MTOR gene mutations upon DNA sequencing. Conclusions We demonstrate a pipeline for the identification and validation of RCC subtype–specific biomarkers that can aid in the confirmation of cell of origin and may facilitate accurate classification and diagnosis of renal tumors. Patient summary FOXI1, RHCG, and LINC01187 are lineage-specific signature genes for chromophobe renal cell carcinoma.
- Published
- 2019
12. Proteogenomic insights into the biology and treatment of HPV-negative head and neck squamous cell carcinoma
- Author
-
Tara Skelly, Wen Jiang, Zhen Zhang, Anupriya Agarwal, Amy M. Perou, Olga Potapova, Christopher R. Kinsinger, Matthew A. Wyczalkowski, David J. Clark, Shuang Cai, Felipe da Veiga Leprevost, Linda Hannick, Chen Huang, Paul D. Piehowski, John McGee, Marcin J. Domagalski, Dmitris Placantonakis, Jianbo Pan, Dana R. Valley, Zhiao Shi, Hui Yin Chang, Karen A. Ketchum, Charles A. Goldthwaite, Małgorzata Wierzbicka, Karsten Krug, Yvonne Shutack, Sara R. Savage, Matthew L. Anderson, Alyssa Charamut, Chandan Kumar-Sinha, Sanford P. Markey, Ratna R. Thangudu, Weiping Ma, Oliver F. Bathe, Antonio Colaprico, Yuxing Liao, Eric E. Schadt, Tomasz Czernicki, Seungyeul Yoo, Xi Chen, Stacey Gabriel, Karl R. Clauser, Daniel C. Rohrer, Uma Borate, Uma Velvulou, Larisa Polonskaya, M. Harry Kane, Dmitry M. Avtonomov, Boris Reva, Jacob J. Day, Barbara Hindenach, Matthew J. Ellis, Katherine A. Hoadley, Emek Demir, Rebecca I. Montgomery, Ewa P. Malc, Fengchao Yu, Lijun Yao, Maciej Wiznerowicz, Annette Marrero-Oliveras, Wojciech Szopa, Sailaja Mareedu, Galen Hostetter, Liqun Qi, Hui Zhang, Yige Wu, David N. Hayes, Shankha Satpathy, Corbin D. Jones, Michael J. Birrer, Xinpei Yi, Nathan Edwards, Fei Ding, Jiang Qian, Ning Qu, Alicia Francis, Daniel Cui Zhou, Jakub Stawicki, Bing Zhang, Rodrigo Vargas Eguez, Tao Liu, Dave Tabor, Maureen Dyer, Brian J. Druker, Gilbert S. Omenn, Azra Krek, Meenakshi Anurag, Melissa Borucki, Mathangi Thiagarajan, Shirley Tsang, Shakti Ramkissoon, Alexey I. Nesvizhskii, Li Ding, Lyubomir Valkov Vasilev, Yifat Geffen, James Suh, Tatiana S. Ermakova, Kakhaber Zaalishvili, Adel K. El-Naggar, Ki Sung Um, Ana I. Robles, Wen-Wei Liang, Richard D. Smith, Pei Wang, Emily S. Boja, Anna Calinawan, Yingwei Hu, Jiayi Ji, Renata Ferrarotto, Hongwei Liu, Jonathan T. Lei, Ramani B. Kothadia, Yize Li, Chelsea J. Newton, Anna Malovannaya, Steven A. Carr, Sandra Cerda, Yuriy Zakhartsev, Stephanie De Young, Eric J. Jaehnig, Peter B. McGarvey, Yan Shi, David I. Heiman, Joseph C. Dort, Karin D. Rodland, Lili Blumenberg, Michael A. Gillette, Piotr A. Mieczkowski, Pankaj Vats, Chet Birger, Yongchao Dou, David Fenyö, Saravana M. Dhanasekaran, Pushpa Hariharan, Eunkyung An, Jeffrey R. Whiteaker, George Miles, Jan Lubinski, Shayan C. Avanessian, Samuel H. Payne, Amanda G. Paulovich, Dmitry Rykunov, Lyudmila Petrenko, Martin Hyrcza, Guo Ci Teo, Alissa M. Weaver, D. R. Mani, Houston Culpepper, Meghan C. Burke, Daniel W. Chan, Bo Wen, Nicollette Maunganidze, Elie Traer, Darlene Tansil, Simona Migliozzi, Luciano Garofano, Qing Kay Li, Donghui Tan, Lori J. Sokoll, Mehdi Mesri, Karna Robinson, Fulvio D'Angelo, Kimberly Elburn, Alexander R. Pico, Umut Ozbek, Michael Schnaubelt, Gad Getz, Francesca Petralia, Andrew G. Sikora, Kai Li, Elena V. Ponomareva, Arul M. Chinnaiyan, Robert Zelt, Jun Zhu, Midie Xu, Dimitar Dimitrov Pazardzhikliev, Negin Vatanian, Grace Zhao, Thomas F. Westbrook, Kyung-Cho Cho, Yuefan Wang, Jason E. McDermott, Jeffrey W. Tyner, William Bocik, Shilpi Singh, Stephen E. Stein, Nancy Roche, Alicia Karz, Shannon Richey, Tara Hiltke, Michael Vernon, Lijun Chen, Henry Rodriguez, Xiaoyu Song, Elizabeth R. Duffy, Lin S. Chen, Liwei Cao, Shrabanti Chowdhury, Marcin Cieślik, Michael C. Wendl, Scott D. Jewell, and Cristina E. Tognon
- Subjects
Adult ,Male ,Proteomics ,0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Cell ,Biology ,Article ,Young Adult ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Cyclin-dependent kinase ,medicine ,Humans ,Aged ,Proteogenomics ,Aged, 80 and over ,Squamous Cell Carcinoma of Head and Neck ,Papillomavirus Infections ,Phosphoproteomics ,Immunotherapy ,Middle Aged ,medicine.disease ,Head and neck squamous-cell carcinoma ,Immune checkpoint ,ErbB Receptors ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female - Abstract
We present a proteogenomic study of 108 human papilloma virus (HPV)-negative head and neck squamous cell carcinomas (HNSCCs). Proteomic analysis systematically catalogs HNSCC-associated proteins and phosphosites, prioritizes copy number drivers, and highlights an oncogenic role for RNA processing genes. Proteomic investigation of mutual exclusivity between FAT1 truncating mutations and 11q13.3 amplifications reveals dysregulated actin dynamics as a common functional consequence. Phosphoproteomics characterizes two modes of EGFR activation, suggesting a new strategy to stratify HNSCCs based on EGFR ligand abundance for effective treatment with inhibitory EGFR monoclonal antibodies. Widespread deletion of immune modulatory genes accounts for low immune infiltration in immune-cold tumors, whereas concordant upregulation of multiple immune checkpoint proteins may underlie resistance to anti-programmed cell death protein 1 monotherapy in immune-hot tumors. Multi-omic analysis identifies three molecular subtypes with high potential for treatment with CDK inhibitors, anti-EGFR antibody therapy, and immunotherapy, respectively. Altogether, proteogenomics provides a systematic framework to inform HNSCC biology and treatment.
- Published
- 2021
13. Integrated Proteogenomic Characterization across Major Histological Types of Pediatric Brain Cancer
- Author
-
Matthew E. Monroe, Saravana M. Dhanasekaran, Brian R. Rood, Zeynep H. Gümüş, Jena Lilly, Samuel G. Winebrake, Richard G. Ivey, William Bocik, Mahdi Sarmady, Alicia Francis, Lamiya Tauhid, Nathan Edwards, Lizabeth Katsnelson, Rui Zhao, Matilda Broberg, Jo Lynne Rokita, Mateusz Koptyra, Henry Rodriguez, Cassie Kline, Shrabanti Chowdhury, Nicole Tignor, Ying Wang, Christopher R. Kinsinger, Antonio Colaprico, Amanda G. Paulovich, Weiping Ma, Emily S. Boja, Tara Hiltke, Sabine Mueller, Liang-Bo Wang, Javad Nazarian, Marcin J. Domagalski, Karl K. Weitz, Jessica B. Foster, Robert Lober, Carina A. Leonard, Bo Zhang, Gerald A. Grant, Anna Calinawan, Gonzalo Lopez, Shuang Cai, Joanna J. Phillips, Guo Ci Teo, July E. Palma, Felipe da Veiga Leprevost, Yiran Guo, Angela Waanders, Xiaoyu Song, Li Ding, Allison Heath, Steven P. Gygi, Rosalie K. Chu, Vasileios Stathias, Bailey Farrow, Oren J. Becher, Dmitry Rykunov, Nithin D. Adappa, Ron Firestein, Adam C. Resnick, Marcin Cieślik, Jennifer Mason, D. R. Mani, Selim Kalayci, Boris Reva, Antonio Iavarone, MacIntosh Cornwell, Uliana J. Voytovich, Gabrielle S. Stone, Miguel A. Brown, Jacob J. Kennedy, Tao Liu, Ronald J. Moore, Emily Kawaler, Eric H. Raabe, Marina A. Gritsenko, Valerie Baubet, Francesca Petralia, Maciej Wiznerowicz, Olena Morozova Vaske, Eric E. Schadt, Ian F. Pollack, Arul M. Chinnaiyan, Meghan Connors, Jason E. Cain, Lei Zhao, Matthew A. Wyczalkowski, Nalin Gupta, Bing Zhang, Jiayi Ji, Marilyn M. Li, Samuel Rivero-Hinojosa, Mariarita Santi, Wenke Liu, John Szpyt, Brian Ennis, Alexey I. Nesvizhskii, Joshua M. Wang, Jeffrey P. Greenfield, Sanjukta Guha Thakurta, Hui Yin Chang, Peter B. McGarvey, Xi Chen, Karen A. Ketchum, Stephan C. Schürer, Sarah Leary, Lili Blumenberg, Matthew J. Ellis, Pei Wang, Anna Maria Buccoliero, Karsten Krug, Chiara Caporalini, Gad Getz, David E. Kram, Pichai Raman, Eric M. Jackson, James N. Palmer, Mehdi Mesri, Kelly V. Ruggles, Chunde Li, Jun Zhu, Sonia Partap, Jeffrey R. Whiteaker, Mirko Scagnet, Krutika S. Gaonkar, Azra Krek, Allison M. Morgan, Tatiana Omelchenko, Richard D. Smith, Elizabeth Appert, Karin D. Rodland, Derek Hanson, Phillip B. Storm, Jamie Moon, Vladislav A. Petyuk, Nathan Young, Travis D. Lorentzen, David Fenyö, Angela N. Viaene, Seungyeul Yoo, Yuankun Zhu, Nicholas A Vitanza, Toan Le, Tatiana Patton, and Ana I. Robles
- Subjects
DNA Copy Number Variations ,Computational biology ,Biology ,Proteomics ,Article ,General Biochemistry, Genetics and Molecular Biology ,Ganglioglioma ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Glioma ,medicine ,Humans ,Gene Regulatory Networks ,RNA, Messenger ,Copy-number variation ,Phosphorylation ,Child ,Proteogenomics ,030304 developmental biology ,Medulloblastoma ,0303 health sciences ,Brain Neoplasms ,Genome, Human ,Phosphoproteomics ,Phosphoproteins ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Mutation ,Atypical teratoid rhabdoid tumor ,Neoplasm Grading ,Neoplasm Recurrence, Local ,Transcriptome ,030217 neurology & neurosurgery - Abstract
We report a comprehensive proteogenomics analysis, including whole-genome sequencing, RNA sequencing, and proteomics and phosphoproteomics profiling, of 218 tumors across 7 histological types of childhood brain cancer: low-grade glioma (n = 93), ependymoma (32), high-grade glioma (25), medulloblastoma (22), ganglioglioma (18), craniopharyngioma (16), and atypical teratoid rhabdoid tumor (12). Proteomics data identify common biological themes that span histological boundaries, suggesting that treatments used for one histological type may be applied effectively to other tumors sharing similar proteomics features. Immune landscape characterization reveals diverse tumor microenvironments across and within diagnoses. Proteomics data further reveal functional effects of somatic mutations and copy number variations (CNVs) not evident in transcriptomics data. Kinase-substrate association and co-expression network analysis identify important biological mechanisms of tumorigenesis. This is the first large-scale proteogenomics analysis across traditional histological boundaries to uncover foundational pediatric brain tumor biology and inform rational treatment selection.
- Published
- 2020
14. Identification and Validation of PCAT14 as Prognostic Biomarker in Prostate Cancer
- Author
-
Yashar S. Niknafs, Marcin Cieślik, Arul M. Chinnaiyan, Xiaojun Jing, Bhavna Malik, Lanbo Xiao, Rohit Mehra, Bui Huynh-Hoa, Xiang Zhang, Xuhong Cao, Rohit Malik, Felix Y. Feng, Edward M. Schaeffer, Susan M. Freier, Shuling Guo, Saravana M. Dhanasekaran, Sudhanshu Shukla, Javed Siddiqui, and Ashley E. Ross
- Subjects
0301 basic medicine ,PCA3 ,Cancer Research ,Cancer ,Disease ,In situ hybridization ,Biology ,Bioinformatics ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,3. Good health ,Gene expression profiling ,Management of prostate cancer ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,Cancer research ,medicine ,Gene - Abstract
Rapid advances in the discovery of long noncoding RNAs (lncRNAs) have identified lineage- and cancer-specific biomarkers that may be relevant in the clinical management of prostate cancer (PCa). Here we assembled and analyzed a large RNA-seq dataset, from 585 patient samples, including benign prostate tissue and both localized and metastatic PCa to discover and validate differentially expressed genes associated with disease aggressiveness. We performed Sample Set Enrichment Analysis (SSEA) and identified genes associated with low versus high Gleason score in the RNA-seq database. Comparing Gleason 6 versus 9+ PCa samples, we identified 99 differentially expressed genes with variable association to Gleason grade as well as robust expression in prostate cancer. The top-ranked novel lncRNA PCAT14, exhibits both cancer and lineage specificity. On multivariate analysis, low PCAT14 expression independently predicts for BPFS ( P =.00126), PSS ( P =.0385), and MFS ( P =.000609), with trends for OS as well ( P =.056). An RNA in-situ hybridization (ISH) assay for PCAT14 distinguished benign vs malignant cases, as well as high vs low Gleason disease. PCAT14 is transcriptionally regulated by AR, and endogenous PCAT14 overexpression suppresses cell invasion. Thus, Using RNA-sequencing data we identify PCAT14, a novel prostate cancer and lineage-specific lncRNA. PCAT14 is highly expressed in low grade disease and loss of PCAT14 predicts for disease aggressiveness and recurrence.
- Published
- 2016
- Full Text
- View/download PDF
15. Characterizing the Therapeutic Potential of a Potent BET Degrader in Merkel Cell Carcinoma
- Author
-
Jonathan Gurkan, Fengyun Su, Kristin M. Juckette, Rohit Malik, Marcin Cieślik, Andrzej A. Dlugosz, Rui Wang, Xuhong Cao, Monique Verhaegen, Xiaojun Jing, Bing Zhou, Sahr Yazdani, Mishaal Yazdani, Jean Tien, Arul M. Chinnaiyan, Jae Eun Choi, Yuping Wang, Paul W. Harms, Ingrid J. Apel, Doris Mangelberger, and Shaomeng Wang
- Subjects
0301 basic medicine ,Cancer Research ,Original article ,BET, bromodomain and extra terminal domain ,Skin Neoplasms ,MCPyV, Merkel cell polyomavirus ,Merkel cell polyomavirus ,chemical and pharmacologic phenomena ,lcsh:RC254-282 ,BET inhibitor ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Hox gene ,Antigens, Viral, Tumor ,Gene ,Polyomavirus Infections ,biology ,Dose-Response Relationship, Drug ,Merkel cell carcinoma ,Chemistry ,Gene Expression Profiling ,Cell Cycle ,Genes, Homeobox ,food and beverages ,Proteins ,hemic and immune systems ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,biology.organism_classification ,Cell Cycle Gene ,Small molecule ,3. Good health ,Carcinoma, Merkel Cell ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer research ,Acetanilides ,Transcriptome ,Heterocyclic Compounds, 3-Ring - Abstract
Studies on the efficacy of small molecule inhibitors in Merkel cell carcinoma (MCC) have been limited and largely inconclusive. In this study, we investigated the therapeutic potential of a potent BET degrader, BETd-246, in the treatment of MCC. We found that MCC cell lines were significantly more sensitive to BETd-246 than to BET inhibitor treatment. Therapeutic targeting of BET proteins resulted in a loss of “MCC signature” genes but not MYC expression as previously described irrespective of Merkel cell polyomavirus (MCPyV) status. In MCPyV+ MCC cells, BETd-246 alone suppressed downstream targets in the MCPyV-LT Ag axis. We also found enrichment of HOX and cell cycle genes in MCPyV− MCC cell lines that were intrinsically resistant to BETd-246. Our findings uncover a requirement for BET proteins in maintaining MCC lineage identity and point to the potential utility of BET degraders for treating MCC.
- Published
- 2018
16. Targeting the MLL complex in castration-resistant prostate cancer
- Author
-
John R. Prensner, Yashar S. Niknafs, Arul M. Chinnaiyan, Nallasivam Palanisamy, Alexey I. Nesvizhskii, Yi-Mi Wu, Dattatreya Mellacheruvu, Xiaojun Jing, Matthew K. Iyer, Pranathi Meda Krishnamurthy, Rohit Malik, Lakshmi P. Kunju, Yuanyuan Qiao, Saravana M. Dhanasekaran, Rachell Stender, Tomasz Cierpicki, Anastasia K. Yocum, Dmitry Borkin, Xuhong Cao, Felix Y. Feng, Amjad Khan, Marcin Cieślik, Xia Jiang, Jolanta Grembecka, Xiaoju Wang, June Escara-Wilke, and Irfan A. Asangani
- Subjects
medicine.drug_class ,Cancer ,General Medicine ,Biology ,Androgen ,medicine.disease ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Androgen receptor ,Leukemia ,Prostate cancer ,hemic and lymphatic diseases ,medicine ,Cancer research ,Myeloid-Lymphoid Leukemia Protein ,Neoplasm ,Signal transduction ,neoplasms - Abstract
Resistance to androgen deprivation therapies and increased androgen receptor (AR) activity are major drivers of castration-resistant prostate cancer (CRPC). Although prior work has focused on targeting AR directly, co-activators of AR signaling, which may represent new therapeutic targets, are relatively underexplored. Here we demonstrate that the mixed-lineage leukemia protein (MLL) complex, a well-known driver of MLL fusion-positive leukemia, acts as a co-activator of AR signaling. AR directly interacts with the MLL complex via the menin-MLL subunit. Menin expression is higher in CRPC than in both hormone-naive prostate cancer and benign prostate tissue, and high menin expression correlates with poor overall survival of individuals diagnosed with prostate cancer. Treatment with a small-molecule inhibitor of menin-MLL interaction blocks AR signaling and inhibits the growth of castration-resistant tumors in vivo in mice. Taken together, this work identifies the MLL complex as a crucial co-activator of AR and a potential therapeutic target in advanced prostate cancer.
- Published
- 2015
17. Abstract 4091: Evaluating the efficacy of a STING agonist in a murine model of prostate cancer
- Author
-
Jonathan Gurkan, Jae Eun Choi, Jean Tien, Marcin Cieślik, Luigi Franchi, and Arul M. Chinnaiyan
- Subjects
Cancer Research ,Oncology - Abstract
Despite the promise of checkpoint inhibitor therapy, prostate cancer has remained resistant to this treatment. Innate immune agonists, however, have been shown to have anti-tumor effects in various cancer types, such as melanoma and colon cancer, and have been nominated to be used in combination with approved anti-PD1/PD-L1 drugs. For instance, STING agonist + anti-PD-1 combination therapy has been demonstrated in a syngeneic mouse model of melanoma. Other innate immune agonists, such as TLR7/8 and TLR9 have also been shown to have potential anti-tumor effects. We hypothesized that innate immune agonists may be effective in the MycCaP syngeneic mouse model of prostate cancer. To compare the efficacy of three innate agonists targeting the STING (3’3’ cGAMP), TLR7/8 (CL097), and TLR9 (ODN2395) pathways, FVB mice were first injected bilaterally with MycCaP subcutaneous tumors. Upon establishment of tumors, mice were administered 3 doses of a single-sided intratumoral injection of each drug. The injected tumors had responses of 50% (6/12), 23% (3/13), and 8% (1/13) for the STING, TLR7/8 and, TLR8 agonists, respectively. Contralateral tumors showed no significant regression. Published data have reported inherent resistance of MycCaP tumors to anti-PD-1 treatment in vivo. Given our results, we hypothesize that the addition of a STING agonist could enhance efficacy of anti-PD-1 therapy in this model. Mice with MycCaP tumors were administered anti-PD1 alone (n=10) or in combination with bilateral injections of the STING agonist (n=14). Combined therapy resulted in significant reduction in tumor size (71%) compared to anti-PD1 alone (9%). Additionally, there was a significant increase in the Ifnb1 (p=.04), Ifng (p=.008), Tnf-α (p=.006), IRF3 (p=.003) and II6 (p Citation Format: Jonathan Gurkan, Jae Eun Choi, Jean Tien, Marcin Cieślik, Luigi Franchi, Arul M. Chinnaiyan. Evaluating the efficacy of a STING agonist in a murine model of prostate 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 4091.
- Published
- 2019
18. Bubble-seq analysis of the human genome reveals distinct chromatin-mediated mechanisms for regulating early- and late-firing origins
- Author
-
Joyce L. Hamlin, Larry D. Mesner, Stefan Bekiranov, Veena Valsakumar, Rebecca R. Pickin, and Marcin Cieślik
- Subjects
Epigenomics ,Transcriptional Activation ,Transcription, Genetic ,Euchromatin ,DNA Replication Timing ,Heterochromatin ,Molecular Sequence Data ,Replication Origin ,Biology ,Cell Line, Tumor ,Genetics ,Deoxyribonuclease I ,Humans ,Genetics (clinical) ,Replication timing ,Models, Genetic ,Genome, Human ,Research ,Sequence Analysis, DNA ,Chromatin ,Genetic Loci ,Origin recognition complex ,Human genome ,HeLa Cells - Abstract
We have devised a method for isolating virtually pure and comprehensive libraries of restriction fragments that contained replication initiation sites (bubbles) in vivo. We have now sequenced and mapped the bubble-containing fragments from GM06990, a near-normal EBV-transformed lymphoblastoid cell line, and have compared origin distributions with a comprehensive replication timing study recently published for this cell line. We find that early-firing origins, which represent ∼32% of all origins, overwhelmingly represent zones, associate only marginally with active transcription units, are localized within large domains of open chromatin, and are significantly associated with DNase I hypersensitivity. Origin “density” falls from early- to mid-S-phase, but rises again in late S-phase to levels only 17% lower than in early S-phase. Unexpectedly, late origin density calculated on the 1-Mb scale increases as a function of increasing chromatin compaction. Furthermore, the median efficiency of origins in late-replicating, heterochromatic domains is only 25% lower than in early-replicating euchromatic loci. Thus, the activation of early- and late-firing origins must be regulated by quintessentially different mechanisms. The aggregate data can be unified into a model in which initiation site selection is driven almost entirely by epigenetic factors that fashion both the long-range and local chromatin environments, with underlying DNA sequence and local transcriptional activity playing only minor roles. Importantly, the comprehensive origin map we have prepared for GM06990 overlaps moderately well with origin maps recently reported for the genomes of four different human cell lines based on the distributions of small nascent strands.
- Published
- 2013
19. MicroRNA-101 regulated transcriptional modulator SUB1 plays a role in prostate cancer
- Author
-
Moloy T. Goswami, A. M. Chinnaiyan, Shannon Carskadon, Stephanie Daignault, Sumit Agarwal, Nallasivam Palanisamy, Sooryanarayana Varambally, Balabhadrapatruni V. S. K. Chakravarthi, Marcin Cieślik, Xiaojun Jing, Darshan S. Chandrashekar, L.P. Kunju, Alyncia D. Robinson, Javed Siddiqui, and Satya S. Pathi
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Biology ,medicine.disease_cause ,Metastasis ,03 medical and health sciences ,Prostate cancer ,Mice ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,EZH2 ,Prostatic Neoplasms ,medicine.disease ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,Gene Knockdown Techniques ,Cancer cell ,Cancer research ,Heterografts ,Proteasome maturation protein ,Original Article ,Carcinogenesis ,Transcription Factors - Abstract
MicroRNA-101, a tumor suppressor microRNA (miR), is often downregulated in cancer and is known to target multiple oncogenes. Some of the genes that are negatively regulated by miR-101 expression include histone methyltransferase EZH2 (enhancer of zeste homolog 2), COX2 (cyclooxygenase-2), POMP (proteasome maturation protein), CERS6, STMN1, MCL-1 and ROCK2, among others. In the present study, we show that miR-101 targets transcriptional coactivator SUB1 homolog (Saccharomyces cerevisiae)/PC4 (positive cofactor 4) and regulates its expression. SUB1 is known to have diverse role in vital cell processes such as DNA replication, repair and heterochromatinization. SUB1 is known to modulate transcription and acts as a mediator between the upstream activators and general transcription machinery. Expression profiling in several cancers revealed SUB1 overexpression, suggesting a potential role in tumorigenesis. However, detailed regulation and function of SUB1 has not been elucidated. In this study, we show elevated expression of SUB1 in aggressive prostate cancer. Knockdown of SUB1 in prostate cancer cells resulted in reduced cell proliferation, invasion and migration in vitro, and tumor growth and metastasis in vivo. Gene expression analyses coupled with chromatin immunoprecipitation revealed that SUB1 binds to the promoter regions of several oncogenes such as PLK1 (Polo-like kinase 1), C-MYC, serine-threonine kinase BUB1B and regulates their expression. Additionally, we observed SUB1 downregulated CDKN1B expression. PLK1 knockdown or use of PLK1 inhibitor can mitigate oncogenic function of SUB1 in benign prostate cancer cells. Thus, our study suggests that miR-101 loss results in increased SUB1 expression and subsequent activation of known oncogenes driving prostate cancer progression and metastasis. This study therefore demonstrates functional role of SUB1 in prostate cancer, and identifies its regulation and potential downstream therapeutic targets of SUB1 in prostate cancer.
- Published
- 2016
20. Abstract 2458: The androgen receptor-regulated lncRNA landscape reveals a role for ARlnc1 in prostate cancer progression
- Author
-
Mats Ljungman, Lisha Wang, Rohit Malik, Yuanyuan Qiao, Yajia Zhang, Jean C. Tien, Marcin Cieślik, Yashar S. Niknafs, Xuhong Cao, Yasuyuki Hosono, Shuling Guo, Hui Jiang, Sethuramasundaram Pitchiaya, Rohit Mehra, and Arul M. Chinnaiyan
- Subjects
0301 basic medicine ,Untranslated region ,Cancer Research ,Gene knockdown ,medicine.drug_class ,Cancer ,Biology ,Androgen ,medicine.disease ,Androgen receptor ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Prostate ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Gene - Abstract
The androgen receptor (AR) signaling plays a key role in the development of the normal prostate as well as prostate cancer. While substantial efforts have been undertaken to study AR-regulated protein-coding genes in primary prostate cancer and castration-resistant prostate cancer, few studies have investigated the role of long noncoding RNAs. In this study, we employed transcriptome sequencing to delineate long noncoding RNAs (lncRNAs) associated with AR signaling in prostate cancer progression. ARlnc1 (AR-regulated lncRNA 1) was identified as being the top AR-induced, cancer-associated lncRNA in an integrative analysis of prostate cancer cell lines and tissues. Not only was ARlnc1 induced by AR, but ARlnc1 also was shown to sustain AR signaling by stabilizing the AR transcript via interaction with the AR 3' UTR. Knockdown of ARlnc1 suppressed AR expression, global AR signaling, and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARlnc1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression, and identifies ARlnc1 as a novel therapeutic target. Citation Format: Yajia Zhang, Sethuramasundaram Pitchiaya, Marcin Cieślik, Yashar S. Niknafs, Jean C. Tien, Yasuyuki Hosono, Lisha Wang, Yuanyuan Qiao, Xuhong Cao, Mats Ljungman, Hui Jiang, Rohit Mehra, Shuling Guo, Rohit Malik, Arul M. Chinnaiyan. The androgen receptor-regulated lncRNA landscape reveals a role for ARlnc1 in prostate cancer progression [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 2458.
- Published
- 2018
21. Inactivation of CDK12 Delineates a Distinct Immunogenic Class of Advanced Prostate Cancer
- Author
-
Yi-Mi Wu, Marcin Cieślik, Arul M. Chinnaiyan, Pankaj Vats, Navonil De Sarkar, Bruce Montgomery, Xuhong Cao, Elisabeth I. Heath, Weiping Zou, Johann S. de Bono, Robert J. Lonigro, Melissa A. Reimers, Felix Y. Feng, Jonathan Chou, Peter S. Nelson, Lisha Wang, Ajjai Alva, Dan R. Robinson, Yu Ning, and Lakshmi P. Kunju
- Subjects
Male ,0301 basic medicine ,DNA Repair ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Mutant ,Mutation, Missense ,Biology ,SPOP ,Genomic Instability ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,RNA, Small Interfering ,Gene ,Neoplasm Staging ,Chemokine CCL21 ,Prostate ,Antibodies, Monoclonal ,Nuclear Proteins ,Prostatic Neoplasms ,Cell cycle ,medicine.disease ,Cyclin-Dependent Kinases ,Immune checkpoint ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Phenotype ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,RNA Interference ,Cancer biomarkers ,Tomography, X-Ray Computed ,Homologous recombination - Abstract
Using integrative genomic analysis of 360 metastatic castration-resistant prostate cancer (mCRPC) samples, we identified a novel subtype of prostate cancer typified by biallelic loss of CDK12 that is mutually exclusive with tumors driven by DNA repair deficiency, ETS fusions, and SPOP mutations. CDK12 loss is enriched in mCRPC relative to clinically localized disease and characterized by focal tandem duplications (FTDs) that lead to increased gene fusions and marked differential gene expression. FTDs associated with CDK12 loss result in highly recurrent gains at loci of genes involved in the cell cycle and DNA replication. CDK12 mutant cases are baseline diploid and do not exhibit DNA mutational signatures linked to defects in homologous recombination. CDK12 mutant cases are associated with elevated neoantigen burden ensuing from fusion-induced chimeric open reading frames and increased tumor T cell infiltration/clonal expansion. CDK12 inactivation thereby defines a distinct class of mCRPC that may benefit from immune checkpoint immunotherapy.
- Published
- 2018
22. The role of entropy and polarity in intermolecular contacts in protein crystals
- Author
-
Zygmunt S. Derewenda and Marcin Cieślik
- Subjects
Models, Molecular ,Databases, Factual ,Protein Conformation ,Entropy ,Crystallography, X-Ray ,Crystal ,chemistry.chemical_compound ,Structural Biology ,Computer Simulation ,Amino Acids ,chemistry.chemical_classification ,Likelihood Functions ,Chemistry ,Intermolecular force ,Proteins ,General Medicine ,Contact order ,Research Papers ,Amino acid ,Crystallography ,Logistic Models ,Monomer ,Models, Chemical ,X-ray crystallography ,Crystallization ,Protein crystallization ,Hydrophobic and Hydrophilic Interactions ,Software ,Entropy (order and disorder) - Abstract
The integrity and X-ray diffraction quality of protein crystals depend on the three-dimensional order of relatively weak but reproducible intermolecular contacts. Despite their importance, relatively little attention has been paid to the chemical and physical nature of these contacts, which are often regarded as stochastic and thus not different from randomly selected protein surface patches. Here, logistic regression was used to analyze crystal contacts in a database of 821 unambiguously monomeric proteins with structures determined to 2.5 A resolution or better. It is shown that the propensity of a surface residue for incorporation into a crystal contact is not a linear function of its solvent-accessible surface area and that amino acids with low exposed surfaces, which are typically small and hydrophobic, have been underestimated with respect to their contact-forming potential by earlier area-based calculations. For any given solvent-exposed surface, small and hydrophobic residues are more likely to be involved in crystal contacts than large and charged amino acids. Side-chain entropy is the single physicochemical property that is most negatively correlated with the involvement of amino acids in crystal contacts. It is also shown that crystal contacts with larger buried surfaces containing eight or more amino acids have cores that are depleted of polar amino acids.
- Published
- 2009
23. Abstractions, Algorithms and Data Structures for Structural Bioinformatics in PyCogent
- Author
-
Cameron Mura, Zygmunt S. Derewenda, and Marcin Cieślik
- Subjects
FOS: Computer and information sciences ,Computer science ,Extensibility ,General Biochemistry, Genetics and Molecular Biology ,Computer Programs ,Computer Science - Software Engineering ,03 medical and health sciences ,Structural bioinformatics ,Computer Science - Data Structures and Algorithms ,Leverage (statistics) ,Data Structures and Algorithms (cs.DS) ,Implementation ,030304 developmental biology ,computer.programming_language ,0303 health sciences ,030302 biochemistry & molecular biology ,Protein structure analysis ,Biomolecules (q-bio.BM) ,Python (programming language) ,File format ,Data structure ,Software Engineering (cs.SE) ,ComputingMethodologies_PATTERNRECOGNITION ,Quantitative Biology - Biomolecules ,FOS: Biological sciences ,Algorithm ,computer - Abstract
To facilitate flexible and efficient structural bioinformatics analyses, new functionality for three-dimensional structure processing and analysis has been introduced into PyCogent -- a popular feature-rich framework for sequence-based bioinformatics, but one which has lacked equally powerful tools for handling stuctural/coordinate-based data. Extensible Python modules have been developed, which provide object-oriented abstractions (based on a hierarchical representation of macromolecules), efficient data structures (e.g. kD-trees), fast implementations of common algorithms (e.g. surface-area calculations), read/write support for Protein Data Bank-related file formats and wrappers for external command-line applications (e.g. Stride). Integration of this code into PyCogent is symbiotic, allowing sequence-based work to benefit from structure-derived data and, reciprocally, enabling structural studies to leverage PyCogent's versatile tools for phylogenetic and evolutionary analyses., Comment: 36 pages, 4 figures (including supplemental information)
- Published
- 2014
- Full Text
- View/download PDF
24. Epigenetic reprogramming in the epithelial-to-mesenchymal transition
- Author
-
Stefan Bekiranov, Lisa G. Gray, Natalya Baranova, Marcin Cieślik, Stephen A. Hoang, Sanjay Chodaparambil, David F. Allison, Jake Wamsley, Manish Kumar, and Marty W. Mayo
- Subjects
Genetics ,Microarray analysis techniques ,lcsh:R ,lcsh:Medicine ,General Medicine ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Chromatin ,Cell biology ,Epigenetic Profile ,Oral Presentation ,lcsh:Q ,Epithelial–mesenchymal transition ,Epigenetics ,lcsh:Science ,Enhancer ,Reprogramming ,Transcription factor - Abstract
The epithelial-to-mesenchymal transition (EMT) is a cellular dedifferentiation process that is critical to development, wound healing and metastasis. Like other cell state transitions, such as differentiation, EMT is accompanied by genome-wide epigenetic reprogramming. However, the relationship between reprogramming and functional changes in the cell is poorly understood. In an A549 non-small cell lung cancer EMT model system we observed changes in chromatin state between epithelial and mesenchymal states. Multivariate analyses were applied to paired (epithelial and mesenchymal) ChIP-seq data for 18 histone modifications/variants and expression microarray data. We observed epigenetic co-regulation of genes associated with EMT, as well as their proximal enhancers. We also observed epigenetic activation or repression of functionally distinct sets of enhancers. These genes and enhancers are regulated and bound by a small set of transcription factors, specifically AP-1, NF-κB and c-Myc. These transcription factors themselves also a show an epigenetic profile similar to the EMT-related genes. Together, these observations suggest a chromatin-mediated transcriptional feedback mechanism that establishes and maintains the phenotypic switch.
- Published
- 2012
25. Abstract 3636: Targeting the MLL complex in castration resistant prostate cancer
- Author
-
Felix Y. Feng, Xuhong Cao, John R. Prensner, Xiaoju Wang, Arul M. Chinnaiyan, Xia Jiang, June Escara-Wilke, Dmitry Borkin, Rohit Malik, Irfan A. Asangani, Amjad Khan, Jolanta Grembecka, Tomasz Cierpicki, Matthew K. Iyer, Marcin Cieślik, Yi-Mi Wu, and Rachell Stender
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Prostate cancer ,business.industry ,Internal medicine ,medicine ,Castration resistant ,medicine.disease ,business - Abstract
Resistance to androgen deprivation therapies and increased androgen receptor (AR) activity are major drivers of castration resistant prostate cancer (CRPC). Substantial prior work has focused on targeting AR directly; however, the identification and therapeutic targeting of co-activators of AR signaling remains an underexplored area of potential clinical significance. Here we demonstrate that the MLL (mixed-lineage leukemia) complex, a well-known contributor in MLL-fusion-positive leukemia, acts as a co-activator of AR signaling. AR directly interacts with the MLL complex via its critical subunit, menin. Small molecule inhibition of the menin-MLL interaction blocks AR signaling and inhibits the growth of castration resistant tumors in vivo. Furthermore, we find that menin is up-regulated in castration resistant prostate cancer and high expression correlates with poor overall survival. Taken together, our study identifies the MLL complex as a critical co-activator of AR that can be targeted in advanced prostate cancer. Citation Format: Rohit Malik, Amjad P. Khan, Irfan A. Asangani, Marcin Cieślik, John R. Prensner, Xiaoju Wang, Matthew K. Iyer, Xia Jiang, Dmitry Borkin, June Escara-Wilke, Rachell Stender, Yi-Mi Wu, Xuhong Cao, Felix Y. Feng, Jolanta Grembecka, Tomasz Cierpicki, Arul M. Chinnaiyan. Targeting the MLL complex in castration resistant prostate cancer. [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 3636. doi:10.1158/1538-7445.AM2015-3636
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.