32 results on '"Jayamanna Wickramasinghe"'
Search Results
2. DAXX-ATRX regulation of p53 chromatin binding and DNA damage response
- Author
-
Nitish Gulve, Chenhe Su, Zhong Deng, Samantha S. Soldan, Olga Vladimirova, Jayamanna Wickramasinghe, Hongwu Zheng, Andrew V. Kossenkov, and Paul. M. Lieberman
- Subjects
Science - Abstract
The tumor suppressor proteins DAXX and ATRX are frequently mutated in cancers with alternative lengthening of telomeres (ALT). This study shows that DAXX-ATRX regulates p53 chromatin accessibility and DNA damage response and that disruption of this pathway is critical for ALT cell survival.
- Published
- 2022
- Full Text
- View/download PDF
3. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy
- Author
-
Rajasekharan Somasundaram, Thomas Connelly, Robin Choi, Hyeree Choi, Anastasia Samarkina, Ling Li, Elizabeth Gregorio, Yeqing Chen, Rohit Thakur, Mohamed Abdel-Mohsen, Marilda Beqiri, Meaghan Kiernan, Michela Perego, Fang Wang, Min Xiao, Patricia Brafford, Xue Yang, Xiaowei Xu, Anthony Secreto, Gwenn Danet-Desnoyers, Daniel Traum, Klaus H. Kaestner, Alexander C. Huang, Denitsa Hristova, Joshua Wang, Mizuho Fukunaga-Kalabis, Clemens Krepler, Fang Ping-Chen, Xiangyang Zhou, Alexis Gutierrez, Vito W. Rebecca, Prashanthi Vonteddu, Farokh Dotiwala, Shashi Bala, Sonali Majumdar, Harsh Dweep, Jayamanna Wickramasinghe, Andrew V. Kossenkov, Jorge Reyes-Arbujas, Kenisha Santiago, Tran Nguyen, Johannes Griss, Frederick Keeney, James Hayden, Brian J. Gavin, David Weiner, Luis J. Montaner, Qin Liu, Lukas Peiffer, Jürgen Becker, Elizabeth M. Burton, Michael A. Davies, Michael T. Tetzlaff, Kar Muthumani, Jennifer A. Wargo, Dmitry Gabrilovich, and Meenhard Herlyn
- Subjects
Science - Abstract
Immune checkpoint therapies (ICT) are promising for treating various cancers, but response rates vary. Here the authors show, in mouse models, that tumor-infiltrating mast cells colocalize with regulatory T cells, coincide with local reduction of MHC-I and CD8 T cells, and is associated with resistance to ICT, which can be reversed by c-kit inhibitor treatment.
- Published
- 2021
- Full Text
- View/download PDF
4. A multi-omics approach to Epstein-Barr virus immortalization of B-cells reveals EBNA1 chromatin pioneering activities targeting nucleotide metabolism.
- Author
-
R Jason Lamontagne, Samantha S Soldan, Chenhe Su, Andreas Wiedmer, Kyoung Jae Won, Fang Lu, Aaron R Goldman, Jayamanna Wickramasinghe, Hsin-Yao Tang, David W Speicher, Louise Showe, Andrew V Kossenkov, and Paul M Lieberman
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Epstein-Barr virus (EBV) immortalizes resting B-lymphocytes through a highly orchestrated reprogramming of host chromatin structure, transcription and metabolism. Here, we use a multi-omics-based approach to investigate these underlying mechanisms. ATAC-seq analysis of cellular chromatin showed that EBV alters over a third of accessible chromatin during the infection time course, with many of these sites overlapping transcription factors such as PU.1, Interferon Regulatory Factors (IRFs), and CTCF. Integration of RNA-seq analysis identified a complex transcriptional response and associations with EBV nuclear antigens (EBNAs). Focusing on EBNA1 revealed enhancer-binding activity at gene targets involved in nucleotide metabolism, supported by metabolomic analysis which indicated that adenosine and purine metabolism are significantly altered by EBV immortalization. We further validated that adenosine deaminase (ADA) is a direct and critical target of the EBV-directed immortalization process. These findings reveal that purine metabolism and ADA may be useful therapeutic targets for EBV-driven lymphoid cancers.
- Published
- 2021
- Full Text
- View/download PDF
5. Comparative transcriptome analysis of endemic and epidemic Kaposi's sarcoma (KS) lesions and the secondary role of HIV-1 in KS pathogenesis.
- Author
-
Salum J Lidenge, Andrew V Kossenkov, For Yue Tso, Jayamanna Wickramasinghe, Sara R Privatt, Owen Ngalamika, John R Ngowi, Julius Mwaiselage, Paul M Lieberman, John T West, and Charles Wood
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
In sub-Saharan Africa, endemic Kaposi's sarcoma (EnKS) is still prevalent despite high incidence of epidemic Kaposi's sarcoma (EpKS) resulting from the on-going HIV-1 epidemic. While KSHV is clearly the etiologic agent of KS, the mechanisms underlying KS development are not fully understood. For example, HIV-1 co-infection and concomitant immune dysfunction have been associated with EpKS development. However, the direct or indirect role(s) of HIV-1, and therefore of immune suppression, in EpKS remains unclear. How, or whether, EpKS is mechanistically distinct from EnKS is unknown. Thus, the absence of HIV-1 co-infection in EnKS provides a unique control for investigating and deciphering whether HIV-1 plays a direct or indirect role in the EpKS tumor microenvironment. We hypothesized that HIV-1 co-infection would induce transcriptome changes that differentiate EpKS from EnKS, thereby defining the direct intra-tumor role of HIV-1 in KS. Comparison of ART-treated and -naïve patients would further define the impact of ART on the KS transcriptome. We utilized RNA-seq followed by multiparameter bioinformatics analysis to compare transcriptomes from KS lesions to uninvolved control skin. We provide the first transcriptomic comparison of EpKS versus EnKS, ART-treated vs-naïve EpKS and male vs female EpKS to define the roles of HIV-1 co-infection, the impact of ART, and gender on KS gene expression profiles. Our findings suggest that ART-use and gender have minimal impact on transcriptome profiles of KS lesions. Gene expression profiles strongly correlated between EpKS and EnKS patients (Spearman r = 0.83, p
- Published
- 2020
- Full Text
- View/download PDF
6. RNA-Seq of Kaposi's sarcoma reveals alterations in glucose and lipid metabolism.
- Author
-
For Yue Tso, Andrew V Kossenkov, Salum J Lidenge, Owen Ngalamika, John R Ngowi, Julius Mwaiselage, Jayamanna Wickramasinghe, Eun Hee Kwon, John T West, Paul M Lieberman, and Charles Wood
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS). It is endemic in a number of sub-Saharan African countries with infection rate of >50%. The high prevalence of HIV-1 coupled with late presentation of advanced cancer staging make KS the leading cancer in the region with poor prognosis and high mortality. Disease markers and cellular functions associated with KS tumorigenesis remain ill-defined. Several studies have attempted to investigate changes of the gene profile with in vitro infection of monoculture models, which are not likely to reflect the cellular complexity of the in vivo lesion environment. Our approach is to characterize and compare the gene expression profile in KS lesions versus non-cancer tissues from the same individual. Such comparisons could identify pathways critical for KS formation and maintenance. This is the first study that utilized high throughput RNA-seq to characterize the viral and cellular transcriptome in tumor and non-cancer biopsies of African epidemic KS patients. These patients were treated anti-retroviral therapy with undetectable HIV-1 plasma viral load. We found remarkable variability in the viral transcriptome among these patients, with viral latency and immune modulation genes most abundantly expressed. The presence of KSHV also significantly affected the cellular transcriptome profile. Specifically, genes involved in lipid and glucose metabolism disorder pathways were substantially affected. Moreover, infiltration of immune cells into the tumor did not prevent KS formation, suggesting some functional deficits of these cells. Lastly, we found only minimal overlaps between our in vivo cellular transcriptome dataset with those from in vitro studies, reflecting the limitation of in vitro models in representing tumor lesions. These findings could lead to the identification of diagnostic and therapeutic markers for KS, and will provide bases for further mechanistic studies on the functions of both viral and cellular genes that are involved.
- Published
- 2018
- Full Text
- View/download PDF
7. Characterizing The Landscape Of Viral Expression In Cancer By Deep Learning.
- Author
-
Abdurrahman Elbasir, Ying Ye, Daniel E. Schäffer, Jayamanna Wickramasinghe, Xue Hao, Paul M. Lieberman, Quaid Morris, Rugang Zhang, Alejandro A. Schäffer, and Noam Auslander
- Published
- 2022
- Full Text
- View/download PDF
8. A distinct core regulatory module enforces oncogene expression in KMT2A-rearranged leukemia
- Author
-
Taku Harada, Yaser Heshmati, Jérémie Kalfon, Monika W. Perez, Juliana Xavier Ferrucio, Jazmin Ewers, Benjamin Hubbell Engler, Andrew Kossenkov, Jana M. Ellegast, Joanna S. Yi, Allyson Bowker, Qian Zhu, Kenneth Eagle, Tianxin Liu, Yan Kai, Joshua M. Dempster, Guillaume Kugener, Jayamanna Wickramasinghe, Zachary T. Herbert, Charles H. Li, Jošt Vrabič Koren, David M. Weinstock, Vikram R. Paralkar, Behnam Nabet, Charles Y. Lin, Neekesh V. Dharia, Kimberly Stegmaier, Stuart H. Orkin, and Maxim Pimkin
- Subjects
Gene Rearrangement ,Leukemia, Myeloid, Acute ,hemic and lymphatic diseases ,Interferon Regulatory Factors ,Genetics ,Humans ,Oncogenes ,Myeloid-Lymphoid Leukemia Protein ,Developmental Biology - Abstract
Acute myeloid leukemia with KMT2A (MLL) rearrangements is characterized by specific patterns of gene expression and enhancer architecture, implying unique core transcriptional regulatory circuitry. Here, we identified the transcription factors MEF2D and IRF8 as selective transcriptional dependencies of KMT2A-rearranged AML, where MEF2D displays partially redundant functions with its paralog, MEF2C. Rapid transcription factor degradation followed by measurements of genome-wide transcription rates and superresolution microscopy revealed that MEF2D and IRF8 form a distinct core regulatory module with a narrow direct transcriptional program that includes activation of the key oncogenes MYC, HOXA9, and BCL2. Our study illustrates a mechanism of context-specific transcriptional addiction whereby a specific AML subclass depends on a highly specialized core regulatory module to directly enforce expression of common leukemia oncogenes.
- Published
- 2022
- Full Text
- View/download PDF
9. Table S2 from A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT
- Author
-
Louise C. Showe, Michael K. Showe, William N. Rom, Brian Nam, Thomas Bauer, Anil Vachani, Sai Yendamuri, Harvey Pass, Jun-Chieh J. Tsay, Gerard Criner, Eric Konnisto, Wen-Hwai Horng, Trisha Kumar, Sandy Widura, Celia Chang, R. Sonali Majumdar, Qin Liu, Jayamanna Wickramasinghe, Noor B. Dawany, Rehman Qureshi, and Andrew V. Kossenkov
- Abstract
Number of samples used on different platforms for training and validation sets
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary File S1 from A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT
- Author
-
Louise C. Showe, Michael K. Showe, William N. Rom, Brian Nam, Thomas Bauer, Anil Vachani, Sai Yendamuri, Harvey Pass, Jun-Chieh J. Tsay, Gerard Criner, Eric Konnisto, Wen-Hwai Horng, Trisha Kumar, Sandy Widura, Celia Chang, R. Sonali Majumdar, Qin Liu, Jayamanna Wickramasinghe, Noor B. Dawany, Rehman Qureshi, and Andrew V. Kossenkov
- Abstract
Ranking of Nanostring Custom panel probes in nPNC classifier
- Published
- 2023
- Full Text
- View/download PDF
11. Figure S2 from A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT
- Author
-
Louise C. Showe, Michael K. Showe, William N. Rom, Brian Nam, Thomas Bauer, Anil Vachani, Sai Yendamuri, Harvey Pass, Jun-Chieh J. Tsay, Gerard Criner, Eric Konnisto, Wen-Hwai Horng, Trisha Kumar, Sandy Widura, Celia Chang, R. Sonali Majumdar, Qin Liu, Jayamanna Wickramasinghe, Noor B. Dawany, Rehman Qureshi, and Andrew V. Kossenkov
- Abstract
Performance of the Pulmonary Nodule Classifier
- Published
- 2023
- Full Text
- View/download PDF
12. Supplemental Methods from Follicle-Stimulating Hormone Receptor Is Expressed by Most Ovarian Cancer Subtypes and Is a Safe and Effective Immunotherapeutic Target
- Author
-
Jose R. Conejo-Garcia, Tyler J. Curiel, Julia Tchou, Denise C. Connolly, Mark G. Cadungog, Qihong Huang, Kiranmai Gumireddy, Shane W. O'Brien, Jenny M. Nguyen, Jayamanna Wickramasinghe, Kyle K. Payne, Amelia J. Tesone, Michael J. Allegrezza, Melanie R. Rutkowski, Nikolaos Svoronos, and Alfredo Perales-Puchalt
- Abstract
Supplemental Methods
- Published
- 2023
- Full Text
- View/download PDF
13. Supplemental Figures and Legends from Follicle-Stimulating Hormone Receptor Is Expressed by Most Ovarian Cancer Subtypes and Is a Safe and Effective Immunotherapeutic Target
- Author
-
Jose R. Conejo-Garcia, Tyler J. Curiel, Julia Tchou, Denise C. Connolly, Mark G. Cadungog, Qihong Huang, Kiranmai Gumireddy, Shane W. O'Brien, Jenny M. Nguyen, Jayamanna Wickramasinghe, Kyle K. Payne, Amelia J. Tesone, Michael J. Allegrezza, Melanie R. Rutkowski, Nikolaos Svoronos, and Alfredo Perales-Puchalt
- Abstract
Supplemental Figure 1. (A) Cytotoxicity of FSHCER or mock transduced T-cells of OVCAR-3 luciferase tumor cells measured by luciferase detection after co-culture of 18 hours. (B) Gating strategy followed for the flow cytometry based cytotoxicity assay. After gating on the singlet cells we could gate the tumor cells and exclude T-cells by FSC and SSC (confirmed by the no Tcell control) and measure the percentage of living cells as double negatives from Annexin-V and 7AAD. Supplemental Figure 2. (A) immunohistochemistry showing a section CAOV3 tumor stained with FSHR18 antibody followed by secondary anti-mouse (FSHR18) or secondary alone (Negative). 20x, scale bar: 1 um. (B) Western blot and qPCR showing FSHR expression of different ovarian cancer and breast cancer tumor cell lines. (C) Tumor volume of three ovarian patient-derived xenograft tumors grown in the flank of NOD-SCID mice (n=2 mice per tumor, one case-one control, single experiment) injected intratumorally with 10 million FSHCER or mock transduced T-cells (arrows mark time of T-cell injection). Common axis used for easier cross comparison. Supplemental Figure 3. (A) FSHR mRNA of FACS-sorted ID8-Defb29/Vegf-a/Fshr from the peritoneal cavity of orthotopic ID8-Defb29/Vegf-a/Fshr-bearing mice treated with either FSHCER, mock transduced T-cells or PBS. (B) Normalized real-time quantitative-PCR of FSHR expression in the human healthy ovarian tissue shown in Figure 3A, a human serous ovarian carcinoma specimen and 2 additional cell lines. (C) Absence of expression of the FSHR in untransduced ID8-Defb29/Vegf-a cells, compared to FSHR-transduced clones (40 PCR cycles).
- Published
- 2023
- Full Text
- View/download PDF
14. Data from Follicle-Stimulating Hormone Receptor Is Expressed by Most Ovarian Cancer Subtypes and Is a Safe and Effective Immunotherapeutic Target
- Author
-
Jose R. Conejo-Garcia, Tyler J. Curiel, Julia Tchou, Denise C. Connolly, Mark G. Cadungog, Qihong Huang, Kiranmai Gumireddy, Shane W. O'Brien, Jenny M. Nguyen, Jayamanna Wickramasinghe, Kyle K. Payne, Amelia J. Tesone, Michael J. Allegrezza, Melanie R. Rutkowski, Nikolaos Svoronos, and Alfredo Perales-Puchalt
- Abstract
Purpose: To define the safety and effectiveness of T cells redirected against follicle-stimulating hormone receptor (FSHR)-expressing ovarian cancer cells.Experimental Design: FSHR expression was determined by Western blotting, immunohistochemistry, and qPCR in 77 human ovarian cancer specimens from 6 different histologic subtypes and 20 human healthy tissues. The effectiveness of human T cells targeted with full-length FSH in vivo was determined against a panel of patient-derived xenografts. Safety and effectiveness were confirmed in immunocompetent tumor-bearing mice, using constructs targeting murine FSHR and syngeneic T cells.Results: FSHR is expressed in gynecologic malignancies of different histologic types but not in nonovarian healthy tissues. Accordingly, T cells expressing full-length FSHR-redirected chimeric receptors mediate significant therapeutic effects (including tumor rejection) against a panel of patient-derived tumors in vivo. In immunocompetent mice growing syngeneic, orthotopic, and aggressive ovarian tumors, fully murine FSHR-targeted T cells also increased survival without any measurable toxicity. Notably, chimeric receptors enhanced the ability of endogenous tumor-reactive T cells to abrogate malignant progression upon adoptive transfer into naïve recipients subsequently challenged with the same tumor. Interestingly, FSHR-targeted T cells persisted as memory lymphocytes without noticeable PD-1–dependent exhaustion during end-stage disease, in the absence of tumor cell immunoediting. However, exosomes in advanced tumor ascites diverted the effector activity of this and other chimeric receptor–transduced T cells away from targeted tumor cells.Conclusions: T cells redirected against FSHR+ tumor cells with full-length FSH represent a promising therapeutic alternative against a broad range of ovarian malignancies, with negligible toxicity even in the presence of cognate targets in tumor-free ovaries. Clin Cancer Res; 23(2); 441–53. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
15. Data from A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT
- Author
-
Louise C. Showe, Michael K. Showe, William N. Rom, Brian Nam, Thomas Bauer, Anil Vachani, Sai Yendamuri, Harvey Pass, Jun-Chieh J. Tsay, Gerard Criner, Eric Konnisto, Wen-Hwai Horng, Trisha Kumar, Sandy Widura, Celia Chang, R. Sonali Majumdar, Qin Liu, Jayamanna Wickramasinghe, Noor B. Dawany, Rehman Qureshi, and Andrew V. Kossenkov
- Abstract
Low-dose CT (LDCT) is widely accepted as the preferred method for detecting pulmonary nodules. However, the determination of whether a nodule is benign or malignant involves either repeated scans or invasive procedures that sample the lung tissue. Noninvasive methods to assess these nodules are needed to reduce unnecessary invasive tests. In this study, we have developed a pulmonary nodule classifier (PNC) using RNA from whole blood collected in RNA-stabilizing PAXgene tubes that addresses this need. Samples were prospectively collected from high-risk and incidental subjects with a positive lung CT scan. A total of 821 samples from 5 clinical sites were analyzed. Malignant samples were predominantly stage 1 by pathologic diagnosis and 97% of the benign samples were confirmed by 4 years of follow-up. A panel of diagnostic biomarkers was selected from a subset of the samples assayed on Illumina microarrays that achieved a ROC-AUC of 0.847 on independent validation. The microarray data were then used to design a biomarker panel of 559 gene probes to be validated on the clinically tested NanoString nCounter platform. RNA from 583 patients was used to assess and refine the NanoString PNC (nPNC), which was then validated on 158 independent samples (ROC-AUC = 0.825). The nPNC outperformed three clinical algorithms in discriminating malignant from benign pulmonary nodules ranging from 6–20 mm using just 41 diagnostic biomarkers. Overall, this platform provides an accurate, noninvasive method for the diagnosis of pulmonary nodules in patients with non–small cell lung cancer.Significance:These findings describe a minimally invasive and clinically practical pulmonary nodule classifier that has good diagnostic ability at distinguishing benign from malignant pulmonary nodules.
- Published
- 2023
- Full Text
- View/download PDF
16. Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidatesfor targeted treatment
- Author
-
Li Chen, Jacqueline Mudd, Michael Ittmann, Carol J. Bult, Amanda R. Kirane, Jelena Randjelovic, Stephen Scott, Yige Wu, Li Ding, Vashisht G. Yennu-Nanda, Jing Wang, Christopher D. Lanier, Maihi Fujita, Emilio Cortes-Sanchez, Sienna Rocha, Susan G. Hilsenbeck, Kian-Huat Lim, Fernanda Martins Rodrigues, Jill Rubinstein, Nicholas Mitsiades, Haiyin Lin, Jayamanna Wickramasinghe, Andrew Butterfield, Bryan E. Welm, Alana L. Welm, Jose P. Zevallos, Jason Held, Nicole B. Coggins, Song Cao, Yuanxin Xi, Brenda C. Timmons, Paul Lott, David Menter, Shunqiang Li, Tina Primeau, Fei Yang, Andrea Wang-Gillam, Ramaswamy Govindan, Dali Li, Brandi Davis-Dusenbery, Sara Seepo, Michael C. Wendl, Jeffrey Grover, Brian S. White, Clifford G. Tepper, Peter N. Robinson, Michael A. Davies, Zhengtao Chu, Michael W. Lloyd, Hua Sun, Xiaoshan Zhang, Tamara Stankovic, Dylan Fingerman, Anuj Srivastava, Luis G. Carvajal-Carmona, Don L. Gibbons, Lijun Yao, Rebecca Aft, Hongyong Zhang, Ismail Meraz, John DiGiovanna, Scott Kopetz, Ling Zhao, Guadalupe Polanco-Echeverry, Feng Chen, Jeremy Hoog, Matthew A. Wyczalkowski, George Xu, John D. Minna, Yi Xu, Julie Belmar, Xiaowei Xu, Luc Girard, Dennis A. Dean, Tijana Borovski, Chong-xian Pan, Cynthia X. Ma, Alexa Morales Arana, Yize Li, Turcin Saridogan, Steven B. Neuhauser, Sandra Scherer, Vicki Chin, Rose Tipton, David R. Gandara, Sherri R. Davies, Argun Akcakanat, Rajesh Patidar, Julie K. Schwarz, Soner Koc, Gao Boning, Michael Kim, Bryce P. Kirby, Yvonne A. Evrard, Hyunsil Park, Christian Frech, Chia-Kuei Mo, Ran Zhang, Brian A. Van Tine, Jonathan W. Reiss, Min Xiao, Xing Yi Woo, Tiffany Le, Ana Estrada, Xiaofeng Zheng, Jeffrey A. Moscow, Mourad Majidi, Nadezhda V. Terekhanova, Katherine Fuh, Erkan Yuca, Timothy A. Yap, Jianhua Zhang, Matthew J. Ellis, Shannon Westin, James H. Doroshow, Vito W. Rebecca, Moon S. Chen, Coya Tapia, Reyka G Jayasinghe, Jack A. Roth, Jithesh Augustine, Ryan C. Fields, Michae T. Tetzlaff, Michael T. Lewis, Kurt W. Evans, Ralph W. deVere White, Brian J. Sanderson, May Cho, Jeffrey H. Chuang, Tiffany Wallace, Ryan Jeon, Ted Toal, Matthew H. Bailey, Bert W. O'Malley, Katherine L. Nathanson, Qin Liu, Benjamin J. Raphael, Jingqin Luo, Salma Kaochar, Huiqin Chen, Rajasekharan Somasundaram, Daniel Cui Zhou, John F. DiPersio, Andrew V. Kossenkov, Bingliang Fang, Vanessa Jensen, Simone Zaccaria, Alexey Sorokin, Ai-Hong Ma, Sidharth V. Puram, Min Jin Ha, Meenhard Herlyn, R. Jay Mashl, Kelly Gale, Bingbing Dai, Lacey E. Dobrolecki, Chieh-Hsiang Yang, and Funda Meric-Bernstam
- Subjects
endocrine system ,Science ,Druggability ,General Physics and Astronomy ,Genomics ,Computational biology ,Biology ,Genome ,digestive system ,General Biochemistry, Genetics and Molecular Biology ,Article ,Research community ,Multiple time ,medicine ,Cancer genomics ,Cancer models ,Tumor xenograft ,Multidisciplinary ,Cancer ,General Chemistry ,medicine.disease ,Pharmacogenomics ,Data integration ,hormones, hormone substitutes, and hormone antagonists - Abstract
Development of candidate cancer treatments is a resource-intensive process, with the research community continuing to investigate options beyond static genomic characterization. Toward this goal, we have established the genomic landscapes of 536 patient-derived xenograft (PDX) models across 25 cancer types, together with mutation, copy number, fusion, transcriptomic profiles, and NCI-MATCH arms. Compared with human tumors, PDXs typically have higher purity and fit to investigate dynamic driver events and molecular properties via multiple time points from same case PDXs. Here, we report on dynamic genomic landscapes and pharmacogenomic associations, including associations between activating oncogenic events and drugs, correlations between whole-genome duplications and subclone events, and the potential PDX models for NCI-MATCH trials. Lastly, we provide a web portal having comprehensive pan-cancer PDX genomic profiles and source code to facilitate identification of more druggable events and further insights into PDXs’ recapitulation of human tumors., Patient-derived xenograft models (PDX) have been extensively used to study the molecular and clinical features of cancers. Here the authors present a cohort of 536 PDX models from 25 cancers, as well as their genomic and evolutionary profiles and their suitability for clinical trials.
- Published
- 2021
17. A deep learning approach reveals unexplored landscape of viral expression in cancer
- Author
-
Abdurrahman Elbasir, Ying Ye, Daniel E. Schäffer, Xue Hao, Jayamanna Wickramasinghe, Konstantinos Tsingas, Paul M. Lieberman, Qi Long, Quaid Morris, Rugang Zhang, Alejandro A. Schäffer, and Noam Auslander
- Abstract
About 15% of human cancer cases are attributed to viral infections. To date, virus expression in tumor tissues has been mostly studied by aligning tumor RNA sequencing reads to databases of known viruses. To allow identification of divergent viruses and rapid characterization of the tumor virome, we develop viRNAtrap, an alignment-free pipeline to identify viral reads and assemble viral contigs. We utilize viRNAtrap, which is based on a deep learning model trained to discriminate viral RNAseq reads, to explore viral expression in cancers and apply it to 14 cancer types from The Cancer Genome Atlas (TCGA). Using viRNAtrap, we uncover expression of unexpected and divergent viruses that have not previously been implicated in cancer and disclose human endogenous viruses whose expression is associated with poor overall survival. The viRNAtrap pipeline provides a way forward to study viral infections associated with different clinical conditions.
- Published
- 2022
- Full Text
- View/download PDF
18. DAXX-ATRX-H3.3 Regulation of p53 Chromatin Binding and DNA Damage Response
- Author
-
Samantha S. Soldan, Nitish Gulve, Olga Vladimirova, Jayamanna Wickramasinghe, Paul M. Lieberman, Hongwu Zheng, Zhong Deng, and Andrew V. Kossenkov
- Subjects
Death-associated protein 6 ,DNA damage ,Chemistry ,Chromatin binding ,ATRX ,Cell biology - Abstract
DAXX and ATRX are tumor suppressor proteins that form a histone H3.3 chaperone complex and are frequently mutated in cancers with the alternative lengthening of telomeres (ALT), such as pediatric glioblastoma. Rapid loss of function of either DAXX or ATRX are not by themselves sufficient to induce the ALT phenotype. However, cells lacking DAXX or ATRX can be readily selected for ALT-like features. Here, we show that DAXX and ATRX null glioblastoma cells with ALT-like features have defects in p53 chromatin binding and DNA damage response regulation. RNA-seq analysis of DAXX or ATRX null U87 glioblastoma cells with ALT-like features revealed that p53 pathway is among perturbed. ALT-selected DAXX and ATRX-null cells had aberrant response to DNA damaging agent etoposide. Both DAXX and ATRX-null ALT cells showed a loss of p53 binding at a subset of response elements. Complementation of DAXX null cells with a wild-type DAXX transgene rescued p53 binding and transcription, while the tumor associated mutation L130R that disrupts ATRX binding was incapable of rescuing p53 chromatin binding. We show that histone H3.3 binding is reduced in DAXX-null cells especially at subtelomeric p53 binding sites and telomere repeats. These findings indicate that DAXX and ATRX function to enable p53 chromatin binding through modulation of histone H3.3 binding, especially at sub-telomeric sites.
- Published
- 2021
- Full Text
- View/download PDF
19. A distinct core regulatory module enforces oncogene expression in KMT2A-rearranged leukemia
- Author
-
Jayamanna Wickramasinghe, Kim Stegmaier, Monika Perez, Jeremie Kalfon, Yaser Heshmati, Ferrucio Jx, Joshua M. Dempster, Behnam Nabet, Chengtao Li, Maxim Pimkin, Qian Zhu, Zachary T. Herbert, Guillaume Kugener, Joanna S. Yi, Neekesh V. Dharia, Charles Y. Lin, Andrew V. Kossenkov, Kenneth Eagle, Taku Harada, Jost Vrabic Koren, David M. Weinstock, Stuart H. Orkin, Vikram R Paralkar, Bowker A, Jazmin Ewers, and Jana M. Ellegast
- Subjects
Mef2 ,KMT2A ,biology ,Transcription (biology) ,biology.protein ,Myeloid leukemia ,Computational biology ,IRF8 ,Gene ,Transcription factor ,Chromatin - Abstract
SummaryA small set of lineage-restricted transcription factors (TFs), termed core regulatory circuitry (CRC), control cell identity and malignant transformation. Here, we integrated gene dependency, chromatin architecture and TF perturbation datasets to characterize 31 core TFs in acute myeloid leukemia (AML). Contrary to a widely accepted model, we detected a modular CRC structure with hierarchically organized, partially redundant and only sparsely interconnected modules of core TFs controlling distinct genetic programs. Rapid TF degradation followed by measurement of genome-wide transcription rates revealed that core TFs directly regulate dramatically fewer genes than previously assumed. Leukemias carrying KMT2A (MLL) rearrangements depend on the IRF8/MEF2 axis to directly enforce expression of the key oncogenes MYC, HOXA9 and BCL2. Our datasets provide an evolving model of CRC organization in human cells, and a resource for further inquiries into and therapeutic targeting of aberrant transcriptional circuits in cancer.
- Published
- 2021
- Full Text
- View/download PDF
20. A Gene Expression Classifier from Whole Blood Distinguishes Benign from Malignant Lung Nodules Detected by Low-Dose CT
- Author
-
Noor Dawany, Gerard J. Criner, Wenhwai Horng, Qin Liu, Thomas L. Bauer, William N. Rom, Rehman Qureshi, Sandy Widura, Andrew V. Kossenkov, Jayamanna Wickramasinghe, Eric Konnisto, Sai Yendamuri, Celia Chang, Anil Vachani, Michael K. Showe, Brian Nam, Trisha Kumar, Louise C. Showe, R. Sonali Majumdar, Harvey I. Pass, and Jun-Chieh J. Tsay
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Article ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Biomarkers, Tumor ,Carcinoma ,medicine ,Humans ,Low dose ct ,Prospective Studies ,Prospective cohort study ,Gene ,Aged ,Whole blood ,Lung ,Microarray analysis techniques ,business.industry ,Gene Expression Profiling ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Multiple Pulmonary Nodules ,Female ,Radiology ,Tomography, X-Ray Computed ,business ,Algorithms - Abstract
Low-dose CT (LDCT) is widely accepted as the preferred method for detecting pulmonary nodules. However, the determination of whether a nodule is benign or malignant involves either repeated scans or invasive procedures that sample the lung tissue. Noninvasive methods to assess these nodules are needed to reduce unnecessary invasive tests. In this study, we have developed a pulmonary nodule classifier (PNC) using RNA from whole blood collected in RNA-stabilizing PAXgene tubes that addresses this need. Samples were prospectively collected from high-risk and incidental subjects with a positive lung CT scan. A total of 821 samples from 5 clinical sites were analyzed. Malignant samples were predominantly stage 1 by pathologic diagnosis and 97% of the benign samples were confirmed by 4 years of follow-up. A panel of diagnostic biomarkers was selected from a subset of the samples assayed on Illumina microarrays that achieved a ROC-AUC of 0.847 on independent validation. The microarray data were then used to design a biomarker panel of 559 gene probes to be validated on the clinically tested NanoString nCounter platform. RNA from 583 patients was used to assess and refine the NanoString PNC (nPNC), which was then validated on 158 independent samples (ROC-AUC = 0.825). The nPNC outperformed three clinical algorithms in discriminating malignant from benign pulmonary nodules ranging from 6–20 mm using just 41 diagnostic biomarkers. Overall, this platform provides an accurate, noninvasive method for the diagnosis of pulmonary nodules in patients with non–small cell lung cancer. Significance: These findings describe a minimally invasive and clinically practical pulmonary nodule classifier that has good diagnostic ability at distinguishing benign from malignant pulmonary nodules.
- Published
- 2019
- Full Text
- View/download PDF
21. Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy
- Author
-
Michela Perego, Hyeree Choi, Mohamed Abdel-Mohsen, Sonali Majumdar, Marilda Beqiri, Fang Wang, Daniel Traum, Gwenn Danet-Desnoyers, Jorge Reyes-Arbujas, Meenhard Herlyn, Frederick Keeney, Clemens Krepler, Elizabeth Gregorio, Prashanthi Vonteddu, Mizuho Fukunaga-Kalabis, Robin Choi, Luis J. Montaner, Johannes Griss, Thomas Connelly, Michael A. Davies, Anastasia Samarkina, Jayamanna Wickramasinghe, Jennifer A. Wargo, Michael T. Tetzlaff, Dmitry I. Gabrilovich, Denitsa Hristova, Shashi Bala, Tran Nguyen, Xue Yang, Alexis Gutierrez, Min Xiao, Rajasekharan Somasundaram, Klaus H. Kaestner, Joshua Wang, James Hayden, Yeqing Chen, Karuppiah Muthumani, Elizabeth M. Burton, Farokh Dotiwala, Anthony Secreto, Xiaowei Xu, Qin Liu, David B. Weiner, Fang Ping-Chen, Lukas Peiffer, Brian J. Gavin, Patricia Brafford, Ling Li, Andrew V. Kossenkov, Kenisha Santiago, Harsh Dweep, Xiangyang Zhou, Meaghan Kiernan, Alexander C. Huang, Jürgen C. Becker, Vito W. Rebecca, and Rohit Thakur
- Subjects
0301 basic medicine ,Chemokine ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,CD34 ,Medizin ,Drug Resistance ,General Physics and Astronomy ,Transgenic ,Mice ,0302 clinical medicine ,Sunitinib ,Medicine ,2.1 Biological and endogenous factors ,Lymphocytes ,Mast Cells ,Aetiology ,Melanoma ,Immune Checkpoint Inhibitors ,Cancer ,B-Lymphocytes ,Multidisciplinary ,biology ,FOXP3 ,Mast cell ,Acquired immune system ,Human Fetal Tissue ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine.drug ,Science ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Vaccine Related ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,parasitic diseases ,Animals ,Humans ,Tumor-Infiltrating ,business.industry ,General Chemistry ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,biology.protein ,Cancer research ,Neoplasm ,Immunization ,business ,CD8 - Abstract
Anti-PD-1 therapy is used as a front-line treatment for many cancers, but mechanistic insight into this therapy resistance is still lacking. Here we generate a humanized (Hu)-mouse melanoma model by injecting fetal liver-derived CD34+ cells and implanting autologous thymus in immune-deficient NOD-scid IL2Rγnull (NSG) mice. Reconstituted Hu-mice are challenged with HLA-matched melanomas and treated with anti-PD-1, which results in restricted tumor growth but not complete regression. Tumor RNA-seq, multiplexed imaging and immunohistology staining show high expression of chemokines, as well as recruitment of FOXP3+ Treg and mast cells, in selective tumor regions. Reduced HLA-class I expression and CD8+/Granz B+ T cells homeostasis are observed in tumor regions where FOXP3+ Treg and mast cells co-localize, with such features associated with resistance to anti-PD-1 treatment. Combining anti-PD-1 with sunitinib or imatinib results in the depletion of mast cells and complete regression of tumors. Our results thus implicate mast cell depletion for improving the efficacy of anti-PD-1 therapy.
- Published
- 2021
22. Comparative transcriptome analysis of endemic and epidemic Kaposi’s sarcoma (KS) lesions and the secondary role of HIV-1 in KS pathogenesis
- Author
-
For Yue Tso, Salum J. Lidenge, Julius Mwaiselage, Andrew V. Kossenkov, Owen Ngalamika, John R. Ngowi, Jayamanna Wickramasinghe, Paul M. Lieberman, Sara R. Privatt, John T. West, and Charles E. Wood
- Subjects
RNA viruses ,Male ,Gene Expression ,HIV Infections ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Transcriptome ,Pathogenesis ,Kaposi Sarcoma ,Medical Conditions ,Immunodeficiency Viruses ,Medicine and Health Sciences ,Biology (General) ,0303 health sciences ,Coinfection ,030302 biochemistry & molecular biology ,Sarcoma ,Genomics ,Kaposi's Sarcoma-Associated Herpesvirus ,Middle Aged ,Infectious Diseases ,Oncology ,Medical Microbiology ,Viral Pathogens ,Viruses ,Herpesvirus 8, Human ,Female ,medicine.symptom ,Pathogens ,Transcriptome Analysis ,Research Article ,Adult ,Herpesviruses ,Patients ,QH301-705.5 ,Immunology ,Inflammation ,Biology ,Microbiology ,03 medical and health sciences ,Young Adult ,Immune system ,Signs and Symptoms ,Virology ,Retroviruses ,medicine ,Genetics ,Humans ,Kaposi's sarcoma-associated herpesvirus ,Molecular Biology ,Gene ,Microbial Pathogens ,Sarcoma, Kaposi ,030304 developmental biology ,Tumor microenvironment ,Gene Expression Profiling ,Lentivirus ,Organisms ,Biology and Life Sciences ,HIV ,Computational Biology ,Cancers and Neoplasms ,RC581-607 ,Genome Analysis ,Health Care ,Co-Infections ,HIV-1 ,Lesions ,Parasitology ,Immunologic diseases. Allergy ,Clinical Medicine ,Carcinogenesis ,DNA viruses - Abstract
In sub-Saharan Africa, endemic Kaposi’s sarcoma (EnKS) is still prevalent despite high incidence of epidemic Kaposi’s sarcoma (EpKS) resulting from the on-going HIV-1 epidemic. While KSHV is clearly the etiologic agent of KS, the mechanisms underlying KS development are not fully understood. For example, HIV-1 co-infection and concomitant immune dysfunction have been associated with EpKS development. However, the direct or indirect role(s) of HIV-1, and therefore of immune suppression, in EpKS remains unclear. How, or whether, EpKS is mechanistically distinct from EnKS is unknown. Thus, the absence of HIV-1 co-infection in EnKS provides a unique control for investigating and deciphering whether HIV-1 plays a direct or indirect role in the EpKS tumor microenvironment. We hypothesized that HIV-1 co-infection would induce transcriptome changes that differentiate EpKS from EnKS, thereby defining the direct intra-tumor role of HIV-1 in KS. Comparison of ART-treated and -naïve patients would further define the impact of ART on the KS transcriptome. We utilized RNA-seq followed by multiparameter bioinformatics analysis to compare transcriptomes from KS lesions to uninvolved control skin. We provide the first transcriptomic comparison of EpKS versus EnKS, ART-treated vs–naïve EpKS and male vs female EpKS to define the roles of HIV-1 co-infection, the impact of ART, and gender on KS gene expression profiles. Our findings suggest that ART-use and gender have minimal impact on transcriptome profiles of KS lesions. Gene expression profiles strongly correlated between EpKS and EnKS patients (Spearman r = 0.83, p, Author summary Despite improved antiretroviral therapy (ART) coverage, Kaposi’s sarcoma (KS) remains the most common cancer in people living with HIV/AIDS. KSHV is known to cause KS, but there are no preventive or curative vaccines. Our understanding of the interactions between KSHV, host cell and the microenvironment is lacking. HIV-1 co-infection has been implicated in KS pathogenesis, but its mechanistic role is unclear. We analyzed transcriptomes from lesions and uninvolved control skin from HIV-1-positive and -negative KS patients to determine the direct or indirect role of HIV-1 in KS development. Transcriptomes from the uninvolved control skin from KS subjects were indistinguishable from that of non-KS healthy individuals. This validates the use of uninvolved control skin to control for gene expression pattern in KS lesions. Despite high concordance in gene expression profiles between HIV-1-positive and -negative KS patients, a subset of genes involved in tumorigenesis and inflammation/immune responses showed higher magnitude of dysregulation in EnKS than EpKS patients. The trend toward lower magnitude of gene dysregulation in EpKS coupled with the absence of HIV-1-transcripts in EpKS suggests an indirect or systemic effect of HIV-1 to promote KS tumorigenesis and possibly explain the high KS incidence in regions with endemic KSHV and HIV-1 co-infections.
- Published
- 2020
23. ADAR1 controls apoptosis of stressed cells by inhibiting Staufen1-mediated mRNA decay
- Author
-
Kazuko Nishikura, Andrew V. Kossenkov, Chunzi Song, Hsin Yao Tang, David W. Speicher, Emmanuel Skordalakes, Hiromitsu Ota, Jayamanna Wickramasinghe, Louise C. Showe, Masayuki Sakurai, and Yusuke Shiromoto
- Subjects
0301 basic medicine ,Gene isoform ,RNA editing ,Adenosine Deaminase ,MAP Kinase Signaling System ,Alu ,RNA Stability ,dsRNA ,Apoptosis ,RNA-binding protein ,Biology ,Article ,stress ,03 medical and health sciences ,0302 clinical medicine ,mRNA decay ,Stress, Physiological ,Structural Biology ,ADAR1 ,Humans ,Phosphorylation ,Molecular Biology ,Exportin-5 ,Kinase ,SMD ,RNA-Binding Proteins ,RNA ,Staufen1 ,Molecular biology ,Cell biology ,Cytoskeletal Proteins ,RNA silencing ,030104 developmental biology ,Cytoplasm ,MAP kinase ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
Both p150 and p110 isoforms of ADAR1 convert adenosine to inosine in double-stranded RNA (dsRNA). ADAR1p150 suppresses the dsRNA-sensing mechanism that activates MDA5-MAVS-IFN signaling in the cytoplasm. In contrast, the biological function of the ADAR1p110 isoform, which is usually located in the nucleus, is largely unknown. Here, we show that stress-activated phosphorylation of ADAR1p110 by MKK6-p38-MSK MAP kinases promotes its binding to Exportin-5 and its export from the nucleus. After translocating to the cytoplasm, ADAR1p110 suppresses apoptosis in stressed cells by protecting many antiapoptotic gene transcripts that contain 3'-untranslated-region dsRNA structures primarily comprising inverted Alu repeats. ADAR1p110 competitively inhibits binding of Staufen1 to the 3'-untranslated-region dsRNAs and antagonizes Staufen1-mediated mRNA decay. Our study reveals a new stress-response mechanism in which human ADAR1p110 and Staufen1 regulate surveillance of a set of mRNAs required for survival of stressed cells.
- Published
- 2017
- Full Text
- View/download PDF
24. Systematic Establishment of Robustness and Standards in Patient-Derived Xenograft Experiments and Analysis
- Author
-
Anuj Srivastava, Shunqiang Li, Brian A. Van Tine, Christian Frech, Carol J. Bult, Rajesh Patidar, Vito W. Rebecca, Jeffrey S. Morris, Michael Davies, Huiqin Chen, Sasi Arunachalam, Jeffrey A. Moscow, Ramaswamy Govindan, Jayamanna Wickramasinghe, Zi-Ming Zhao, James H. Doroshow, Adam Stanojevic, Dennis A. Dean, Funda Meric-Bernstam, Jacqueline Rosains, Michael T. Lewis, Bryan E. Welm, Min Xiao, Jelena Randjelovic, Sherri R. Davies, Lily Chen, Brandi N. Davis-Dusenbery, David A. Nix, Meenhard Herlyn, Li Ding, Jack DiGiovanna, Xing Yi Woo, Jack A. Roth, Min Jin Ha, Steven B. Neuhauser, Ryan Jeon, Peter N. Robinson, Bingliang Fang, Michael Lloyd, Tamara Stankovic, Jeffrey H. Chuang, Yvonne A. Evrard, Nevena Miletic, Alana L. Welm, Isheeta Seth, and Andrew V. Kossenkov
- Subjects
endocrine system ,0303 health sciences ,Computer science ,Cancer ,Computational biology ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Robustness (computer science) ,030220 oncology & carcinogenesis ,medicine ,In patient ,030304 developmental biology - Abstract
Patient-Derived Xenografts (PDXs) are tumor-in-mouse models for cancer. PDX collections, such as those supported by the NCI PDXNet program, are powerful resources for preclinical therapeutic testing. However, variations in experimental design and analysis procedures have limited interpretability. To determine the robustness of PDX studies, the PDXNet tested temozolomide drug response for three pre-validated PDX models (sensitive, resistant, and intermediate) across four blinded PDX Development and Trial Centers (PDTCs) using independently selected SOPs. Each PDTC was able to correctly identify the sensitive, resistant, and intermediate models, and statistical evaluations were concordant across all groups. We also developed and benchmarked optimized PDX informatics pipelines, and these yielded robust assessments across xenograft biological replicates. These studies show that PDX drug responses and sequence results are reproducible across diverse experimental protocols. Here we share the range of experimental procedures that maintained robustness, as well as standardized cloud-based workflows for PDX exome-seq and RNA-Seq analysis and for evaluating growth.
- Published
- 2019
- Full Text
- View/download PDF
25. Author Correction: Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts
- Author
-
Michael Lloyd, R. Jay Mashl, Yvonne A. Evrard, Jeffrey A. Moscow, Jong Il Kim, Alana L. Welm, Michael A. Davies, Carol J. Bult, Jayamanna Wickramasinghe, Rania El Botty, Dennis A. Dean, Jessica Giordano, Anuj Srivastava, Sandra D. Scherer, Jacqueline Rosains, Christian Frech, Elisabetta Marangoni, Jeffrey H. Chuang, Ryan Jeon, Shunqiang Li, Matthew H. Bailey, Yun-Suhk Suh, Elodie Modave, Yuanxin Xi, Enzo Medico, Li Ding, Livio Trusolino, Adam Lafferty, Vito W. Rebecca, Han-Kwang Yang, Jing Wang, Annette T. Byrne, Xing Yi Woo, Alice C. O’Farrell, Claudio Isella, Li Chen, Brandi N. Davis-Dusenbery, James H. Doroshow, Sherri R. Davies, Diether Lambrechts, Jos Jonkers, Bingliang Fang, Charles Lee, Roebi de Bruijn, Violeta Serra, Jack A. Roth, Rajesh Patidar, Funda Meric-Bernstam, Petra ter Brugge, Andrew V. Kossenkov, Hyun-Soo Kim, Andrea Bertotti, Emilio Cortes-Sanchez, Chieh-Hsiang Yang, Ramaswamy Govindan, Francesco Galimi, Jelena Randjelovic, Zi-Ming Zhao, Bryan E. Welm, Hua Sun, Meenhard Herlyn, and Michael T. Lewis
- Subjects
Oncology ,medicine.medical_specialty ,DNA Copy Number Variations ,MEDLINE ,Biology ,Polymorphism, Single Nucleotide ,Mice ,Text mining ,Internal medicine ,Databases, Genetic ,Exome Sequencing ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Author Correction ,Cancer ,business.industry ,Published Erratum ,medicine.disease ,Xenograft Model Antitumor Assays ,Computational biology and bioinformatics ,Gene Expression Regulation, Neoplastic ,business - Abstract
Patient-derived xenografts (PDXs) are resected human tumors engrafted into mice for preclinical studies and therapeutic testing. It has been proposed that the mouse host affects tumor evolution during PDX engraftment and propagation, affecting the accuracy of PDX modeling of human cancer. Here, we exhaustively analyze copy number alterations (CNAs) in 1,451 PDX and matched patient tumor (PT) samples from 509 PDX models. CNA inferences based on DNA sequencing and microarray data displayed substantially higher resolution and dynamic range than gene expression-based inferences, and they also showed strong CNA conservation from PTs through late-passage PDXs. CNA recurrence analysis of 130 colorectal and breast PT/PDX-early/PDX-late trios confirmed high-resolution CNA retention. We observed no significant enrichment of cancer-related genes in PDX-specific CNAs across models. Moreover, CNA differences between patient and PDX tumors were comparable to variations in multiregion samples within patients. Our study demonstrates the lack of systematic copy number evolution driven by the PDX mouse host.
- Published
- 2021
- Full Text
- View/download PDF
26. Author Correction: CTCF interacts with the lytic HSV-1 genome to promote viral transcription
- Author
-
Sheryl T. Smith, Nigel W. Fraser, Fengchao Lang, Chunfu Zheng, Olga Vladimirova, Jayamanna Wickramasinghe, Xin Li, Yu Xiao, Lihong Li, Vikrant Singh, Jumin Zhou, Qihan Li, Guijun Chen, Hongbo Han, Danfeng Lu, Paul M. Lieberman, and Benxia Hu
- Subjects
Genetics ,Multidisciplinary ,Lytic cycle ,Transcription (biology) ,CTCF ,Science ,Medicine ,HSL and HSV ,Biology ,Genome - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2021
27. A multi-omics approach to Epstein-Barr virus immortalization of B-cells reveals EBNA1 chromatin pioneering activities targeting nucleotide metabolism
- Author
-
Hsin-Yao Tang, Chenhe Su, Paul M. Lieberman, Jayamanna Wickramasinghe, Louise C. Showe, Aaron R. Goldman, Andreas Wiedmer, David W. Speicher, R. Jason Lamontagne, Fang Lu, Samantha S. Soldan, Andrew V. Kossenkov, and Kyoung-Jae Won
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,B Cells ,Gene Expression ,Biochemistry ,Epigenesis, Genetic ,White Blood Cells ,Adenosine deaminase ,Animal Cells ,Transcription (biology) ,hemic and lymphatic diseases ,Gene expression ,Medicine and Health Sciences ,Biology (General) ,B-Lymphocytes ,0303 health sciences ,biology ,Nucleotides ,Chromosome Biology ,030302 biochemistry & molecular biology ,Genomics ,Chromatin ,Cell biology ,Host-Pathogen Interactions ,Metabolome ,Purine Metabolism ,Epigenetics ,Metabolic Pathways ,Cellular Types ,Transcriptome Analysis ,Reprogramming ,Research Article ,Cell Physiology ,QH301-705.5 ,Immune Cells ,Immunology ,Microbiology ,Viral Proteins ,03 medical and health sciences ,Virology ,DNA-binding proteins ,Genetics ,Humans ,Gene Regulation ,Antibody-Producing Cells ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Blood Cells ,Biology and Life Sciences ,Computational Biology ,Proteins ,Cell Biology ,RC581-607 ,Cell Transformation, Viral ,Genome Analysis ,Regulatory Proteins ,Metabolism ,Epstein-Barr Virus Nuclear Antigens ,CTCF ,biology.protein ,Parasitology ,Cell Immortalization ,Immunologic diseases. Allergy ,Transcriptome ,Transcription Factors ,Interferon regulatory factors - Abstract
Epstein-Barr virus (EBV) immortalizes resting B-lymphocytes through a highly orchestrated reprogramming of host chromatin structure, transcription and metabolism. Here, we use a multi-omics-based approach to investigate these underlying mechanisms. ATAC-seq analysis of cellular chromatin showed that EBV alters over a third of accessible chromatin during the infection time course, with many of these sites overlapping transcription factors such as PU.1, Interferon Regulatory Factors (IRFs), and CTCF. Integration of RNA-seq analysis identified a complex transcriptional response and associations with EBV nuclear antigens (EBNAs). Focusing on EBNA1 revealed enhancer-binding activity at gene targets involved in nucleotide metabolism, supported by metabolomic analysis which indicated that adenosine and purine metabolism are significantly altered by EBV immortalization. We further validated that adenosine deaminase (ADA) is a direct and critical target of the EBV-directed immortalization process. These findings reveal that purine metabolism and ADA may be useful therapeutic targets for EBV-driven lymphoid cancers., Author summary EBV immortalization recapitulates aspects of natural B-cell development from germinal center reaction to memory B-cell differentiation. Here, we provide an integrated multi-omic approach to investigating the EBV-induced changes in B-cell transcriptome using RNA-seq, epigenome by ATAC-seq, and metabolome by mass spectrometry of polar metabolites. We identify purine metabolism as a major pathway that is significantly altered by EBV in each data set. We focus on EBNA1 and find that it binds directly to enhancer elements in a subset of genes, including key regulators of purine metabolism such as adenosine deaminase (ADA) and adenylate kinase 4 (AK4). EBV immortalized B-cells are strongly dependent on ADA for proliferation and inhibitors of ADA prevent B-cell immortalization by EBV. These findings identify purine metabolism as a major pathway targeted by EBV during primary infection and suggest that EBNA1 functions as a pioneering transcription factor that reprograms key regulatory genes in purine metabolism pathway.
- Published
- 2021
- Full Text
- View/download PDF
28. BET Inhibitors Suppress ALDH Activity by Targeting ALDH1A1 Super-Enhancer in Ovarian Cancer
- Author
-
David W. Speicher, Jayamanna Wickramasinghe, Katherine C. Palozola, Rugang Zhang, Hengrui Zhu, Alessandro Gardini, Benjamin G. Bitler, Louise C. Showe, Anil K. Sood, Kenneth S. Zaret, Qin Liu, James E. Bradner, Xiangfan Yin, Zhiguo Zhang, Sherry Y. Wu, Tamas Ordog, Yuhki Yokoyama, Jose R. Conejo-Garcia, Jeong Heon Lee, and Andrew V. Kossenkov
- Subjects
0301 basic medicine ,Cancer Research ,BRD4 ,Aldehyde dehydrogenase ,Cell Cycle Proteins ,Enhancer RNAs ,Carcinoma, Ovarian Epithelial ,Article ,Aldehyde Dehydrogenase 1 Family ,BET inhibitor ,Mice ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasms, Glandular and Epithelial ,Ovarian Neoplasms ,biology ,Nuclear Proteins ,Retinal Dehydrogenase ,Cancer ,Azepines ,Aldehyde Dehydrogenase ,Triazoles ,medicine.disease ,Bromodomain ,ALDH1A1 ,030104 developmental biology ,Oncology ,biology.protein ,Cancer research ,Female ,Cisplatin ,Ovarian cancer ,Transcription Factors - Abstract
The emergence of tumor cells with certain stem-like characteristics, such as high aldehyde dehydrogenase (ALDH) activity due to ALDH1A1 expression, contributes to chemotherapy resistance and tumor relapse. However, clinically applicable inhibitors of ALDH activity have not been reported. There is evidence to suggest that epigenetic regulation of stem-related genes contributes to chemotherapy efficacy. Here, we show that bromodomain and extraterminal (BET) inhibitors suppress ALDH activity by abrogating BRD4-mediated ALDH1A1 expression through a super-enhancer element and its associated enhancer RNA. The clinically applicable small-molecule BET inhibitor JQ1 suppressed the outgrowth of cisplatin-treated ovarian cancer cells both in vitro and in vivo. Combination of JQ1 and cisplatin improved the survival of ovarian cancer–bearing mice in an orthotopic model. These phenotypes correlate with inhibition of ALDH1A1 expression through a super-enhancer element and other stem-related genes in promoter regions bound by BRD4. Thus, targeting the BET protein BRD4 using clinically applicable small-molecule inhibitors, such as JQ1, is a promising strategy for targeting ALDH activity in epithelial ovarian cancer. Cancer Res; 76(21); 6320–30. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
29. Frontline Science: Microbiota reconstitution restores intestinal integrity after cisplatin therapy
- Author
-
Nikolaos Svoronos, Michael J. Allegrezza, Kyle K. Payne, Jessica A. Mine, Alfredo Perales-Puchalt, Subir Biswas, Tara Lee Costich, Logan M. Nickels, Melanie R. Rutkowski, Ricardo A. Chaurio, Jairo Perez-Sanz, Jayamanna Wickramasinghe, Carmen M. Anadon, Joseph Calmette, and Jose R. Conejo-Garcia
- Subjects
0301 basic medicine ,medicine.drug_class ,Immunology ,Antibiotics ,Antineoplastic Agents ,Biology ,Systemic inflammation ,03 medical and health sciences ,0302 clinical medicine ,Intestinal mucosa ,Ruminococcus gnavus ,medicine ,Tumor Cells, Cultured ,Immunology and Allergy ,Animals ,Peritoneal Neoplasms ,Cisplatin ,Ovarian Neoplasms ,Mucin ,Cell Biology ,Fecal Microbiota Transplantation ,medicine.disease ,Intestinal epithelium ,Gastrointestinal Microbiome ,Intestines ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Dysbiosis ,Female ,medicine.symptom ,medicine.drug - Abstract
Due to their cytotoxic activities, many anticancer drugs cause extensive damage to the intestinal mucosa and have antibiotic activities. Here, we show that cisplatin induces significant changes in the repertoire of intestinal commensal bacteria that exacerbate mucosal damage. Restoration of the microbiota through fecal-pellet gavage drives healing of cisplatin-induced intestinal damage. Bacterial translocation to the blood stream is correspondingly abrogated, resulting in a significant reduction in systemic inflammation, as evidenced by decreased serum IL-6 and reduced mobilization of granulocytes. Mechanistically, reversal of dysbiosis in response to fecal gavage results in the production of protective mucins and mobilization of CD11b+ myeloid cells to the intestinal mucosa, which promotes angiogenesis. Administration of Ruminococcus gnavus, a bacterial strain selectively depleted by cisplatin treatment, could only partially restore the integrity of the intestinal mucosa and reduce systemic inflammation, without measurable increases in the accumulation of mucin proteins. Together, our results indicate that reconstitution of the full repertoire of intestinal bacteria altered by cisplatin treatment accelerates healing of the intestinal epithelium and ameliorates systemic inflammation. Therefore, fecal microbiota transplant could paradoxically prevent life-threatening bacteremia in cancer patients treated with chemotherapy.
- Published
- 2017
30. FOLLICLE-STIMULATING HORMONE RECEPTOR IS EXPRESSED BY MOST OVARIAN CANCER SUBTYPES AND IS A SAFE AND EFFECTIVE IMMUNOTHERAPEUTIC TARGET
- Author
-
Jose R. Conejo-Garcia, Nikolaos Svoronos, Michael J. Allegrezza, Jayamanna Wickramasinghe, Qihong Huang, Kyle K. Payne, Kiranmai Gumireddy, Amelia J. Tesone, Melanie R. Rutkowski, Shane W. O'Brien, Denise C. Connolly, Julia Tchou, Mark G. Cadungog, Jenny M. Nguyen, Tyler J. Curiel, and Alfredo Perales-Puchalt
- Subjects
0301 basic medicine ,endocrine system ,Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Biology ,Exosomes ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Ovarian Neoplasms ,Cancer ,Ascites ,Immunotherapy ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Immunoediting ,030220 oncology & carcinogenesis ,Immunology ,Receptors, FSH ,Female ,Ovarian cancer ,Follicle-stimulating hormone receptor - Abstract
Purpose: To define the safety and effectiveness of T cells redirected against follicle-stimulating hormone receptor (FSHR)-expressing ovarian cancer cells. Experimental Design: FSHR expression was determined by Western blotting, immunohistochemistry, and qPCR in 77 human ovarian cancer specimens from 6 different histologic subtypes and 20 human healthy tissues. The effectiveness of human T cells targeted with full-length FSH in vivo was determined against a panel of patient-derived xenografts. Safety and effectiveness were confirmed in immunocompetent tumor-bearing mice, using constructs targeting murine FSHR and syngeneic T cells. Results: FSHR is expressed in gynecologic malignancies of different histologic types but not in nonovarian healthy tissues. Accordingly, T cells expressing full-length FSHR-redirected chimeric receptors mediate significant therapeutic effects (including tumor rejection) against a panel of patient-derived tumors in vivo. In immunocompetent mice growing syngeneic, orthotopic, and aggressive ovarian tumors, fully murine FSHR-targeted T cells also increased survival without any measurable toxicity. Notably, chimeric receptors enhanced the ability of endogenous tumor-reactive T cells to abrogate malignant progression upon adoptive transfer into naïve recipients subsequently challenged with the same tumor. Interestingly, FSHR-targeted T cells persisted as memory lymphocytes without noticeable PD-1–dependent exhaustion during end-stage disease, in the absence of tumor cell immunoediting. However, exosomes in advanced tumor ascites diverted the effector activity of this and other chimeric receptor–transduced T cells away from targeted tumor cells. Conclusions: T cells redirected against FSHR+ tumor cells with full-length FSH represent a promising therapeutic alternative against a broad range of ovarian malignancies, with negligible toxicity even in the presence of cognate targets in tumor-free ovaries. Clin Cancer Res; 23(2); 441–53. ©2016 AACR.
- Published
- 2016
31. CTCF interacts with the lytic HSV-1 genome to promote viral transcription
- Author
-
Danfeng Lu, Jumin Zhou, Chunfu Zheng, Vikrant Singh, Lihong Li, Fengchao Lang, Olga Vladimirova, Sheryl T. Smith, Xin Li, Jayamanna Wickramasinghe, Paul M. Lieberman, Benxia Hu, Nigel W. Fraser, Guijun Chen, Hongbo Han, Qihan Li, and Yu Xiao
- Subjects
0301 basic medicine ,CCCTC-Binding Factor ,Transcription, Genetic ,viruses ,RNA polymerase II ,Genome, Viral ,Herpesvirus 1, Human ,Virus Replication ,Article ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Transcription (biology) ,Chlorocebus aethiops ,Animals ,Humans ,Author Correction ,Vero Cells ,Genetics ,Multidisciplinary ,biology ,RNA ,Chromatin Assembly and Disassembly ,Chromatin ,030104 developmental biology ,HEK293 Cells ,chemistry ,Lytic cycle ,Viral replication ,CTCF ,biology.protein ,RNA, Viral ,RNA Polymerase II ,DNA ,HeLa Cells ,Protein Binding - Abstract
CTCF is an essential chromatin regulator implicated in important nuclear processes including in nuclear organization and transcription. Herpes Simplex Virus-1 (HSV-1) is a ubiquitous human pathogen, which enters productive infection in human epithelial and many other cell types. CTCF is known to bind several sites in the HSV-1 genome during latency and reactivation, but its function has not been defined. Here, we report that CTCF interacts extensively with the HSV-1 DNA during lytic infection by ChIP-seq, and its knockdown results in the reduction of viral transcription, viral genome copy number and virus yield. CTCF knockdown led to increased H3K9me3 and H3K27me3, and a reduction of RNA pol II occupancy on viral genes. Importantly, ChIP-seq analysis revealed that there is a higher level of CTD Ser2P modified RNA Pol II near CTCF peaks relative to the Ser5P form in the viral genome. Consistent with this, CTCF knockdown reduced the Ser2P but increased Ser5P modified forms of RNA Pol II on viral genes. These results suggest that CTCF promotes HSV-1 lytic transcription by facilitating the elongation of RNA Pol II and preventing silenced chromatin on the viral genome.
- Published
- 2016
32. Satb1 Overexpression Drives Tumor-Promoting Activities in Cancer-Associated Dendritic Cells
- Author
-
Eva Brencicova, Amelia J. Tesone, Alfredo Perales-Puchalt, Mark E. Borowsky, Jose R. Conejo-Garcia, Jayamanna Wickramasinghe, Andrew V. Kossenkov, Julia Tchou, Gabriel A. Rabinovich, Jenny M. Nguyen, Nikolaos Svoronos, Michael J. Allegrezza, Tom L. Stephen, Kyle K. Payne, and Melanie R. Rutkowski
- Subjects
0301 basic medicine ,Galectin 1 ,Cellular differentiation ,Immune tolerance ,Histones ,purl.org/becyt/ford/1 [https] ,Mice ,0302 clinical medicine ,Conditional gene knockout ,TRANSCRIPTION FACTOR ,RNA, Small Interfering ,Receptor, Notch1 ,Promoter Regions, Genetic ,Mice, Knockout ,Ovarian Neoplasms ,Cell Differentiation ,SATB1 ,Bioquímica y Biología Molecular ,3. Good health ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,030220 oncology & carcinogenesis ,Galectin-1 ,Female ,Signal transduction ,CIENCIAS NATURALES Y EXACTAS ,Protein Binding ,Signal Transduction ,Biology ,DENDRITIC CELLS ,Article ,General Biochemistry, Genetics and Molecular Biology ,Ciencias Biológicas ,03 medical and health sciences ,INFLAMMATION ,Immune Tolerance ,Animals ,Humans ,IMMUNE TOLERANCE ,purl.org/becyt/ford/1.6 [https] ,Transcription factor ,Cell Proliferation ,Interleukin-6 ,RBPJ ,Histocompatibility Antigens Class II ,Dendritic Cells ,Matrix Attachment Region Binding Proteins ,030104 developmental biology ,Immunology ,Neoplasm Transplantation ,Transcription Factors - Abstract
Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression. Fil: Tesone, Amelia J.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Rutkowski, Melanie R.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Brencicova, Eva. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Svoronos, Nikolaos. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Perales Puchal, Alfredo. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Stephen, Tom L.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Allegrezza, Michael J.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Payne, Kyle K.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Nguyen, Jenny M.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos Fil: Wickramasinghe, Jayamanna. The Wistar Institute. Center for Systems and Computational Biology; Estados Unidos Fil: Tchou, Julia. University of Pennsylvania; Estados Unidos Fil: Borowsky, Mark E.. Christiana Care Health System. Helen F. Graham Cancer Center; Estados Unidos Fil: Rabinovich, Gabriel Adrián. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; Argentina Fil: Kossenkov, Andrew V.. The Wistar Institute. Center for Systems and Computational Biology; Estados Unidos Fil: Conejo Garcia, José R.. The Wistar Institute. Tumor Microenvironment and Metastasis Program; Estados Unidos
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.