Research, Research Resources and Publications
Taking a basic science research approach to understanding cancer
Research in the Department of Epigenetics and Molecular Carcinogenesis (EMC) aims to define the mechanisms that control normal cell proliferation, differentiation, survival, and genome maintenance to identify the aberrations in these processes that drive cancer. EMC research focus areas include: Cellular and Molecular Mechanisms of Carcinogenesis; Cancer Genetics and Epigenetics; Genome Integrity - DNA Replication, Recombination and Repair; and Cancer Stem Cells, Apoptosis and Autophagy. EMC hosts research laboratories and research resources on both the North and South MD Anderson campuses in Houston, Texas.
Research
Cellular and Molecular Mechanisms of Carcinogenesis
The overall goal of our carcinogenesis research is to understand the basic cellular and molecular mechanisms that allow the transformation of normal cells into cancer cells. Defining these mechanisms will help identify new cancer targets and novel strategies for identifying, treating, and preventing cancer.
Departmental carcinogenesis research includes:
- Discovering new genetic contributors and drivers of cancers
- Identifying genetic and/or therapeutic vulnerabilities in cancers (e.g. CRISPR screens identifying synthetic lethality)
- Defining genetic, epigenetic, immunologic, and transcriptional alterations accompanying cancer initiation and progression
- Investigating cell signaling pathways involved in cancer induction and progression
Developing novel computational approaches for cancer research
Enormous amounts of data have been generated from high-throughput, genome-wide experimental approaches, thus creating a pressing need for novel computational approaches to mine and interpret complex "-omics" data sets. Understanding these data sets will help define both the players in biological interaction networks and their functions. Research in the department is combining in-silico approaches with wet-lab experiments to identify drivers and attenuators of cancer. Likewise, the development of CRISPR technology has led to new high-throughput approaches such as genome-wide synthetic lethal screens that also benefit from computational approaches, like those being developed in the department to aid the design and interpretation of high-throughput screens.
Implementing cutting-edge animal models?
Our research relies on the development of genetically engineered animal models for investigating the stepwise molecular changes that occur during carcinogenesis, the function of key genes and gene variants in cancer development, and preclinical prevention and therapeutic studies. A number of existing models are being used for these mechanistic studies, including models for skin, mammary gland, prostate, thymus and blood cancers, but we are also creating novel mouse models using modern knockout/knock-in technologies (e.g. CRISPR-Cas9) to study specific genes and pathways involved in cancer induction and progression.?
Making discoveries at the intersection of immunology and tumor biology
Immunotherapy has been hailed as the fourth pillar of cancer therapy joining surgery, chemotherapy, and radiation treatment. Yet, this treatment approach does not work for all patients. Collaborative departmental research has revealed that patients who respond to immunotherapy have antibody responses against their tumors and that the presence of B cells within a tumor may serve as a marker to predict patient immunotherapy response. These findings are leading to new questions about how B cells in tumors affect immunotherapy response.
Examples of faculty members who are defining the Cellular and Molecular Mechanisms of Carcinogenesis:
¡ª Role of WWOX in cancer and disease; genomic determinants controlling development and progression of pre-invasive breast lesions
Shawn Bratton ¡ª Autophagy and prostate cancer; caspase-activating complexes
Charles Ishak ¡ª Repetitive DNA elements and viral mimicry in tumorigenesis and cancer therapy
Kevin McBride ¡ª B cell function and antibody repertoire; B cell lymphoma; role of B cells in immunotherapy response
¡ª Regulation of thymus development and homeostasis; role of thymic epithelial cells in regulating T-cell development and T-cell receptor repertoire selection
Margarida Almeida Santos ¡ª Tumor promoting roles of genome guardians in leukemia
Angela Ting ¡ª Role of microbiome-expressed genes in altering host DNA to drive cancer
Richard Wood ¡ª Role of DNA polymerase theta in genome stability
Han Xu ¡ª Global view of transcriptional and epigenetic regulation using CRISPR screens; machine-learning and statistical algorithms for high-throughput experiments
Cancer Genetics & Epigenetics
Epigenetic factors that regulate DNA methylation, histone modification and chromatin organization can act as either oncogenes or tumor suppressors. Departmental epigenetics research seeks to define both the normal functions of these factors as well as their roles in cancer formation or suppression.
Readers, Writers and Erasers of Epigenetic Marks
Epigenetic marks include cytosine methylation and hydroxymethylation of DNA and methylation, acetylation, phosphorylation, and ubiquitination of histones. These marks are created by enzymes called "writers." Epigenetic "readers" are effector proteins that bear domains that recognize the specific marks left by the "writers," and epigenetic "erasers" can remove these marks. Thus, multiple epigenomes can be created from a single genome.
Uncovering how epigenetic changes and mutations in epigenetic proteins alter gene expression
The major outcome of epigenetic change is alterations in gene expression. Some epigenetic changes are required for normal development and stem cell differentiation, whereas others are aberrant and can lead to diseases, including immunodeficiency, centromeric region instability and facial anomalies syndrome (ICF) and leukemia and other cancers. Importantly, many epigenetic proteins are mutated in human disease, and epigenetic alterations may be just as important as DNA mutations in driving cancer. In addition to controlling gene transcription, these chromatin-modifying enzymes regulate other processes that require access to DNA, including DNA replication and repair.
Departmental epigenetic research areas include:
- Uncovering epigenetic factors controlling developmental reprogramming
- Discovering biological roles of histone and non-histone lysine and arginine methylation
- Defining the structure and function of epigenetic proteins and epigenetic marks
- Finding new cellular functions for ATP-dependent chromatin remodelers
- Identifying and characterizing ¡°readers¡± of epigenetic marks
- Understanding the role of histone modifications in DNA repair
- Deciphering crosstalk between: histone modifications; histone modifications and DNA methylation; and histone modifications and post-translational modifications of non-histone substrates
- Developing technical and computational tools for genome-wide DNA methylation assessment
- Defining the functions of DNA methylases and DNA methylation in both normal development and cancer
- Understanding the role of DNA methylation-regulated alternative polyadenylation in human health and disease
- Defining the contributions of repetitive DNA elements and viral mimicry to cancer initiation and progression and patient therapy response
Epigenetics research provides new avenues for cancer treatment
Epimutations, unlike genetic mutations, can be reversed by chemotherapeutic intervention, which makes epigenetic therapy conceptually appealing. Researchers in the department are screening and identifying small-molecule regulators of epigenetic modifiers and evaluating their potential as anti-cancer drugs, providing clear translational relevance to this research.
Making foundational protein arginine methyltransferase inhibitor discoveries
EMC department members worked with Experimental Radiation Oncology to show that the protein arginine methyltransferase CARM1 might be a useful therapeutic target for small cell lung cancer (SCLC) by demonstrating that both CARM1 and its target nuclear factor I B (NFIB) are required for the rapid onset of cancer in a preclinical SCLC model. Further, they showed that CARM1 inhibition combined with etoposide and cisplatin (EP - the current standard of care treatment) led to partial tumor regression in SCLC patient-derived xenograft models, which was superior to that of either CARM1 inhibition or EP alone.
In another example, departmental researchers combined their expertise to use cell culture experiments, computational approaches evaluating cancer cell line RNAi screens, and animal xenograft models to show that small molecule inhibition of CARM1 in CREBBP/EP300-mutated diffuse large B-cell lymphomas (DLBCLs) reduces histone acetyltransferase activity and causes synthetic lethality through downregulation of CBP-target genes. This synthetic interaction reveals the possibility that combination therapy using CARM1 inhibitors with CBP/p300 inhibitors may be useful for treating DLBCLs and other cancers with non-mutated CREBBP/EP300.
Examples of faculty engaged in epigenetics research:
Blaine Bartholomew ¡ª ATP-dependent chromatin remodelers influence on chromatin dynamics and non-coding RNA transcription
Mark Bedford ¡ª Arginine methylation in cellular processes; development of technologies to identify readers of proteins bearing specific epigenetic marks
Taiping Chen ¡ª Biological function of histone methylases and demethylases in development and disease
Xiaodong Cheng ¡ª Structure and function of readers, writers, and erasers of DNA modifications and their associated histone modifications
Francesca Cole ¡ª Epigenetic regulation of meiotic chromosome organization & pairing
Charles Ishak ¡ª DNA repetitive elements in cancer initiation, development and therapeutic response
Margarida Almeida Santos ¡ª Epigenetic regulators affecting hematopoietic stem cells
Angela Ting ¡ª Defining novel, clinically relevant functions for DNA methylation and the mechanics of DNA methylation in cancer
DNA Replication, Recombination & Repair - Genomic Stability
Understanding how DNA is damaged and repaired is fundamental to cancer research
DNA breaks can result from molecularly programmed, intentional DNA damage or non-programmed, incidental DNA damage. Intentional breaks results from specialized cellular processes such as those needed for accurate segregation of chromosomes during meiosis and for creating immune system diversity. In contrast, incidental DNA damage results from exposure to DNA damaging agents from both external and internal sources. External sources include ultraviolet radiation from the sun and chemicals in the environment. Internal sources include reactive chemical species, such as oxygen and water as well as accidental DNA breaks formed during DNA replication. Moreover, many cancer therapies induce irreparable DNA damage leading to cell death. Therefore, investigating how cells respond to and repair DNA damage is important for understanding the causes of cancer and developing new treatments. Research in this area employs a broad range of approaches including protein biochemistry, single-cell genomics, high-resolution microscopy, and genetically engineered mouse models.
Departmental research related to genomic stability includes:
- Defining molecular mechanisms of DNA double-strand break repair
- Discovering new DNA repair pathways and proteins
- Defining the functions of DNA polymerases in DNA repair
- Characterizing DNA damage and repair processes in normal cell function and in carcinogenesis
- Learning how DNA damage and repair contribute to immune system diversity
- Unraveling the relationships between DNA damage, chromatin remodeling and DNA repair
- Understanding how DNA methylation influences DNA mutation and repair
When DNA damage causes cells to go awry
Department investigators are studying the protein machinery involved in several DNA repair pathways, including homologous recombination and DNA end-joining processes for the repair of DNA double-strand breaks, and nucleotide excision repair for the repair of ultraviolet radiation-induced DNA damage and other strand-distorting lesions.
First mammalian HR precursors identified
EMC members uncovered the first evidence of homologous recombination (HR) precursors in mammals. Through molecular and cellular characterizations of >1 million haploid genome equivalents from 13 independent mouse single and double mutant conditions, two distinct recombination precursors were identified that form during mouse meiosis including a double Holliday junction (dHJ) that requires the MutL homolog MLH3, but not its nuclease activity, to form.
Faculty are also uncovering how programmed DNA damage and repair are involved in normal cellular processes, such as meiosis and immune system B cell development, as well as how these normal processes go awry and contribute to cancer and disease.
Other active areas of research include investigations into the actions of DNA polymerases at repair sites, how chromatin-modifying proteins cooperate with the DNA repair machinery to facilitate repair in the context of chromatin, the mechanisms underlying the conversion of DNA damage into the mutations that cause cancer, and the mechanisms that allow cells to tolerate and survive DNA damage.
Examples of faculty members leading the way in DNA Replication, Recombination, and Repair :
Xiaodong Cheng ¡ª Role of DNA methylation in DNA mutation and repair
Francesca Cole ¡ª DSB repair by homologous recombination during meiosis
Kevin McBride ¡ª Role of programmed DNA damage in creating antibody diversity and leading to lymphomagenesis
Margarida Almeida Santos ¡ª DNA damage-induced differentiation of stem-like cancer cells
Richard Wood ¡ª Nucleotide excision repair, DNA crosslink repair, role of polymerases in DNA damage tolerance
Cancer Stem Cells & Programmed Cell Death
Stem cells are undifferentiated cells that are unique in their ability to self-renew to create more stem cells while also being able to create daughter cells that can differentiate into other cell types. Most, if not all, cancerous states reflect inappropriate or incomplete cellular differentiation. Aggressive, therapy resistant cancer cells often resemble stem cells in terms of their transcription profiles and self-renewal capacities. Likewise, unregulated growth, abnormal cell division, and defective apoptotic?cell death pathways are hallmark features of tumors.
The goal of research in this area is to define normal stem cell biology and developmental pathways as well as to define genetic, molecular, and biochemical mechanisms that regulate cell proliferation, apoptosis and autophagy and relate these pathways to carcinogenesis.
Specific research in this area includes:
- Defining pathways that govern stem cell biology and embryo development
- Understanding DNA damage response in cancer stem cells
- Epigenetic modifications in embryonic, adult, and cancer stem cells
- Epigenetic mechanisms in early embryos and germ cells
- Role of caspases in apoptosis
- Apoptosis and autophagy in normal and disease processes
Using stem cells to learn about cancer
Our research using mouse embryonic and adult stem cells has led to several foundational discoveries. For example, research centered on GCN5, a histone acetyltransferase component of the SAGA complex, uncovered a Myc-SAGA axis that is critical for driving stem cells to pluripotency and is essential for the expression of cell-cycle genes driven by MYC overexpression in a mouse model of B cell lymphoma. Studies of SETDB1, a lysine methyltransferase that deposits the repressive H3K9me3 mark, revealed that SETDB1-dependent gene repression is essential for preserving intestinal stem cell identity by modulating the Wnt and Notch signaling pathways. Other research using mouse models of MLL-rearranged acute myeloid leukemia showed that protein arginine methyltransferase 5 (PRMT5) is essential for the initiation and maintenance of MLL-AF9-mediated leukemia and that inhibiting PRMT5 restores normal differentiation to hematopoietic stem cells.
Apoptosis and autophagy: cancer inhibitors and facilitators
Autophagy (self-cannibalism) and apoptosis (programmed cell death) are fundamental cellular processes that provide mechanisms for cell survival (autophagy) and cell death (apoptosis). Autophagy provides cells with means not only to survive cellular stress and to recycle cellular materials and organelles but also to rid themselves of damaged, malformed, or foreign material. Although autophagy helps suppress carcinogenesis, it can also promote tumor progression, metastasis, and cancer therapy resistance. Apoptosis is required for normal organismal development, but also provides a mechanism to stop cells with DNA damage from dividing. Many chemotherapeutics work by causing DNA damage and inducing apoptosis; however, defects in apoptosis contribute to both tumorigenesis and resistance to cancer treatment. Therefore, defining the molecular mechanisms guiding autophagy and apoptosis, and the pathways allowing cross-talk between them, will provide insights into how cells evade and succumb to cancer treatments.
Faculty with research interests in Cancer Stem Cells and Programmed Cell Death:
Blaine Bartholomew ¡ª Chromatin remodelers in pluripotency and development
Shawn Bratton ¡ª Heat shock- and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis; caspase-activating complexes and the apoptosome; structure and function of autophagosomes; role of autophagy in prostate cancer
Taiping Chen ¡ª Epigenetic regulation of embryonic and adult stem cell behavior and function
Margarida Almeida Santos ¡ª Epigenetic regulation of cancer stem cells
EMC Research Resources
Flow Cytometry - EMC
Flow Cytometry-EMC provides cutting-edge resources to dissect the cellular and molecular mechanisms of developmental and disease processes, by aiding researchers in identifying, isolating and quantifying cell types and/or cell subsets, while also offering individualized training on specific instruments and assisting with data analysis and experimental design.
Although Flow Cytometry-EMC focuses on EMC users, non-departmental users may also be accommodated, as time and resources permit.
For microscopy services formerly provided by the Flow Cytometry and Cellular Imaging Core-EMC, please see the Advanced Microscopy Core. Please also see the MD Anderson, CCSG-supported, full-service Flow Cytometry and Cellular Imaging Core Facility, directed by Michael Andreeff, M.D., Ph.D.
Flow Cytometry-EMC, located in South Campus Research Building 4, provides assisted use services (hourly fee) to researchers for analytical flow cytometry on all instruments, including the BD LSR Fortessa, BD FACSAria? Fusion SORP, BD FACSAria? Fusion and BDAccuri C6PlusTM. Assisted cell sorts are also available on the BD FACSAria? Fusion SORP and BD FACSAria? Fusion. Instrument training is mandatory for users wishing to perform unassisted analyses. The fee schedule is listed in . Please contact Pam Whitney (pjwhitney@mdanderson.org) prior to scheduling initial services.
Our Services
Analytical Flow Cytometry
Flow cytometry is used broadly in cell biological research. Protein expression or specific indicators of cell status can be measured simultaneously for individual cells within a larger, heterogeneous population. Flow Cytometry-EMC provides access to both to advanced instruments and expert staff to meet researcher needs, while specializing in developing complex assays to quantitatively examine cell surface and cytoplasmic antigens, cell cycle, apoptosis, "side populations" and fluorescent transgene expression (e.g. GFP, YFP, RFP and BFP).
Cell Sorting
Flow Cytometry-EMC cell sorting equipment includes a BD FACSAria? Fusion, 17 parameter, high-speed cell sorter for processing of ¡Ü BSL2 samples within a biosafety cabinet and a BD FACSAria? SORP, 18 parameter, high-speed cell sorter. Users can isolate a wide variety of different cell types based on many different combinations of antibody-based stains, fluorescent protein expression, and viability indicators. Cells are detected using BD FACSDiva? software and can be collected from a single heterogeneous sample into 1-4 tubes, simultaneously.
Recent software upgrades allow for index sorting, letting researchers identify the origin of single cells collected from within the population defined by their experimental parameters. The automatic cell-dispensing unit can dispense single cells into 96- or 384-well plates. The sample chamber and collection apparatus are climate controlled and aerosol contained.
Flow Cytometry-EMC also operates a Miltenyi Biotec autoMACS Pro Cell Separator, an immunomagnetic cell separation system that can be used to enrich or deplete a population of cells prior to sorting on one of our BD instruments.
Other Services
- Multi-parameter analysis and sorting
- Full-spectrum analysis of fluorescent proteins
- autoMACSpro available for depletion/enrichment of cell populations prior to flow experiments
- Sorting cells into a variety of tubes and plates including 96- and 384-well plates
- Index sorting allowing identification of which experimental population a particular cell was derived
- Guidance with experimental design
- Consultation and training
First-Time Users and Training
First-time users should contact Pam Whitney (pjwhitney@mdanderson.org) to schedule a consultation before finalizing any experimental plans. This consultation will aid researchers in optimizing their experimental approach, assay design, and fluorescent panels to ensure recovery of meaningful data, and will define user expectations and service costs for the project. Please schedule your appointment at least one week prior to the anticipated date of service to ensure availability of their chosen instrument. Flow personnel can also assist with data analysis using FlowJo software, or they can perform analysis for a specific project, as time permits, for an additional fee.
Instrument training is mandatory for any user who would perform unassisted flow cytometric analyses. Training is provided by Pam Whitney (pjwhitney@mdanderson.org) and requires uninterrupted blocks of time over multiple days. Please note that all users are responsible for archiving their own data. Flow Cytometry-EMC personnel can assist users in learning this practice.
Additional Information and Resources
Sample Preparation
Experimental Samples
- All samples should be at a concentration of 5.0 x 106 cells/ml in appropriate buffer in 12 x 75 FACS tubes or 15 ml conical tubes depending upon instrument.
- Bring additional buffer for concentration adjustments.
- All samples must be filtered before running on the instruments.
Control Samples
- Each experiment must have proper controls. Biological controls for data samples could include isotype controls for antibody staining or stained normal cycling cells for cell cycle analysis.
- The instrument setup requires additional controls: unstained cells and single-stained color controls.
- Contact Pam Whitney (pjwhitney@mdanderson.org) for more information about the requirements for your experiment.
Spectrum Viewer & Laser Configuration Files
Tissue Culture Hints
Contact Us
Ellen Richie, Ph.D.
Facility Director
Professor
Epigenetics and Molecular Carcinogenesis
Phone:?832-750-7181
Email:?erichie@mdanderson.org
Pamela Whitney
Lab Manager
Office Phone:?832-750-7179
Office Location: 4SCR4.1347
Lab Phone: 832-750-7201
Lab Locations:?4SCR4.2026 and 2028
Email:?pjwhitney@mdanderson.org
Quick Links
Our Equipment
BD FACSAria?Fusion SORP
The BD FACSAria?Fusion SORP is a 5-laser, 18-parameter, high-speed cell sorter, with automatic cell-dispensing unit, run by platform personnel. This instrument can be used to isolate and capture viable cell subsets or single cells for molecular, genetic and/or functional analyses. This instrument can distinguish many fluorescent proteins, stem cell and side populations, in addition to fluorescent antibody preparations. Cells are detected by digital BD FACSDiva? software and collected into one or as many as four tubes simultaneously from a single sample. The cell-dispensing unit on this instrument can dispense single cells into 96- or 384-well plates. The sample chamber and collection apparatus are climate-controlled and aerosol-contained. A laser configuration file for this instrument may be downloaded here.
BD FACSAria? Fusion
The BD FACSAria? Fusion is a 5-laser, 17-parameter high-speed cell sorter with biosafety cabinet and automatic cell-dispensing unit. It is the primary flow instrument and is run by platform personnel. It can be used to isolate and capture viable cell subsets or single cells for molecular, genetic, and functional analyses. The instrument provides exceptional sensitivity and resolution for optimal multicolor analysis and cell sorting. It allows users to distinguish many fluorescent proteins, fluorescent antibody preparations, and other markers to identify stem cell and side populations. Cells are detected by digital BD FACSDiva? software and collected into one, or as many as four tubes, simultaneously. The automatic cell-dispensing unit deposits single cells into 96- or 384-well plates. The enhanced stream stability increases sorting purity and efficiency to help isolate minor subsets of cells (e.g. stem cells) for subsequent in vitro and in vivo experiments. This instrument is contained in a bio-safety cabinet permitting analysis of human specimens while ensuring user protection. A laser configuration file for this instrument may be downloaded here.
BD LSRFortessa?
The BD LSRFortessa? is a 4-laser, 15-parameter cell analyzer equipped with digital BD FACSDiva? software. It can be used for quantification of cellular subsets based on the expression of surface or intracellular antigens, analysis of apoptosis and cell cycle parameters, and multiparameter analyses. Individualized training on the BD LSRFortessa? for unassisted operation is available. A laser configuration file for this instrument may be downloaded here.
Miltenyi autoMACS? Pro Separator
The autoMACS? Pro Separator by Miltenyi is an automated high-speed immunomagnetic cell separation system that can process multiple samples at a time. Many cell and tissue types, including whole blood, can be processed with the use of immunomagnetic beads. Cells can be separated using positive selection, depletion, or untouched isolation programs. It can sort more than 10 million cells per second from samples of up to 4¡Á109 total cells. The system reduces manual sample handling and increases reproducibility.
BD Accuri C6Plus? Cell Analyzer
The newest Flow Cytometry-EMC instrument is a compact, easy-to-use, BD Accuri C6PlusTM 2 laser, 4-parameter cell analyzer. It is optimized for fluorchromes such as FITC, PE, PerCP-Cy?5.5, APC, and BD Horizon Brilliant? Blue 515 and can analyze fluorescent proteins such as GFP, YFP, and mCherry.
Bioinformatics and Biostatistics Services - EMC
Bioinformatics
EMC Bioinformatics faculty members work collaboratively with investigators to provide novel computational methods, tools and algorithms for the advancement of cancer disease research. Our bioinformatics faculty engage in collaborative research in the areas of bioinformatics, statistical genetics and systems biology to retrieve and analyze various kinds of biological data including DNA/RNA sequences, protein sequences and structures, microarray data and RT-PCR data for the purpose of facilitating biological discovery. Special emphasis is placed on next-generation sequencing data analysis.
Biostatistics
Biostatistics services are augmented with additional support from faculty in the Quantitative Sciences division. Many modern research projects generate data sets that require statistical analyses. Biostatistics staff members provide expertise for such projects and can aid in early project conception, helping develop a fully elaborated study design, and aiding with data collection and analysis. Early statistical planning and incorporating power calculations into study design can help researchers implement more cost-effective projects by helping determine the appropriate size and scope of a study as well as aiding with approaches for more meaningful data collection and analysis.
Research Areas & Projects
- Developing working relationships with MD Anderson research faculty and external collaborators to expand the use of advanced computational techniques and statistical analyses, especially as related to next-generation sequencing (e.g. RNA-Seq, DNA-Seq, ChIP-Seq) and DNA methylation analysis
- Providing guidance to faculty, students and coworkers in the use of specialized applications, programming, computation and statistical analysis techniques for interpretation of biological data
- Conducting computational biology and bioinformatics/biostatistics seminars and workshops for researchers and students
- Co-developing or contributing to grant proposals with research faculty, professional colleagues and external collaborators to apply advanced computing and related bioinformatics research to the study of human cancers and disease
- Maintaining awareness of emerging computational technologies and new statistical methods, especially to work with large data sets
Contact Us
Co-leaders
Bin Liu, Ph.D.?
Professor
Epigenetics and Molecular Carcinogenesis
BLiu1@MDAnderson.org
Yue Lu, Ph.D.?
Associate Professor
Epigenetics and Molecular Carcinogenesis
Lu4@MDAnderson.org
Staff
Hai (Kevin) Lin, M.S.
Senior Statistical Analyst
Epigenetics and Molecular Carcinogenesis
KeLin@MDAnderson.org
Recombinant Antibody Creation and Production Platform
The McBride Lab hosts the Recombinant Antibody Production and Creation Platform (RAPCP). The platform is used to develop custom, recombinant, monoclonal antibodies using a propriety method for isolating single-antigen-specific plasma cells, following inoculation into mice.
Curated antibody-encoding genes are cloned into expression vectors for recombinant antibody production and purification. The antibodies can then be tested in collaboration with the end-user, using a number of assays based on user priorities. The process results in sequence-defined, recombinant antibodies in as few as 30-90 days. For more information, please see the McBride Lab Research page.
Epigenomics Profiling Core (EpiCore)
The Epigenomics Profiling Core (EpiCore) is an institutional resource for epigenomics research that brings scientific expertise, advanced assays, and specialized equipment not otherwise readily available to MD Anderson Cancer Center researchers. The core offers cost-effective, full-service protocols to investigate DNA methylation (Pyrosequencing Methylation Analysis, RRBS, cfMeDIP-seq, WGBS), chromatin accessibility (ATAC-Seq), and DNA-protein interactions (ChIP-Seq and CUT&RUN). This core facilitates researchers¡¯ efforts to define epigenetic mechanisms that alter chromatin and regulate transcription in normal and disease conditions. To learn more, please see the Epigenomics Profiling Core website.
Advanced Microscopy Core (AMC)
The Advanced Microscopy Core enables any number of studies including those centered on cellular protein distribution, cell fate, tumor heterogeneity, drug delivery and tumoricidal activity, DNA damage repair, development, neurodegeneration, cell migration/invasion, angiogenesis and cancer-stroma interactions. Staff can guide users through tissue fixation, permeabilization, vibratome sectioning, and tissue clearing to optimize specific imaging needs and develop pilot applications for investigators. Staff will assist in selecting the optimal instrument, fuorophores, and fluorophore combinations for a user¡¯s particular study, as well as helping users apply image processing software, such as the IMARIS 4-D Image Analysis and Tracking software, for their downstream analyses.
Please see the Advanced Microscopy Core website for more information.
AMC Instruments include:
- DeltaVision OMX Blaze V4 Super-Resolution Microscope
- Leica TCS SP8 Confocal Microscope with Environmental Chamber
- Zeiss LSM880 Confocal Microscope with Airyscan
- Leica SP8 DIVE Multiphoton (2-Photon) Microscope
- Zeiss Gaussian Lightsheet 7 (LS7) Microscope
- LifeCanvas SmartSPIM Lightsheet Microscope
- Zeiss Axio Observer Z1
- INCell Analyzer 6500 HS Slide/Plate Scanner
Selected Publications
2025
Ishak CA, Marhon SA, Tchrakian N, Hodgson A, Loo Yau H, Gonzaga IM, Peralta M, Lungu IM, Gomez S, Liang SB, Shen SY, Chen R, Chen J, Chatterjee B, Wanniarachchi KN, Lee J, Zehrbach N, Hosseini A, Mehdipour P, Sun S, Solovyov A, Ettayebi I, Francis KE, He A, Wu T, Feng S, da Silva Medina T, Campos de Almeida F, Bayani J, Li J, MacDonald S, Wang Y, Garcia SS, Arthofer E, Diab N, Srivastava A, Austin PT, Sabatini PJB, Greenbaum BD, O'Brien CA, Shepherd TG, Tsao MS, Chiappinelli KB, Oza AM, Clarke BA, Rottapel R, Lheureux S, De Carvalho DD. Cancer Discov. 2025 Jan 7. doi: 10.1158/2159-8290.CD-24-0094. Online ahead of print. PMID: 39776167
2024
Carvajal-Maldonado D, Li Y, Returan M, Averill AM, Doubli¨¦ S, Wood RD. J Biol Chem. 2024 Jul;300(7):107461.
Chen YC, Bhaskara GB, Lu Y, Lin K, Dent SYR. Genes Dev. 2024 Sep 19;38(15-16):738-754.
Deogharia M, Venegas-Zamora L, Agrawal A, Shi M, Jain AK, McHugh KJ, Altamirano F, Marian AJ, Gurha P. Cardiovasc Res. 2024 May 7;120(6):630-643.
Du Z, Lin M, Li Q, Guo D, Xue Y, Liu W, Shi H, Chen T, Dan J. J Cell Physiol. 2024 Sep;239(9):e31337.
Fijen C, Drogalis Beckham L, Terino D, Li Y, Ramsden DA, Wood RD, Doubli¨¦ S, Rothenberg E. Mol Cell. 2024 Apr 18;84(8):1460-1474.e6.
Hardikar S, Ren R, Ying Z, Zhou J, Horton JR, Bramble MD, Liu B, Lu Y, Liu B, Coletta LD, Shen J, Dan J, Zhang X, Cheng X, Chen T. Sci Adv. 2024 Aug 23;10(34):eadr0036.
Hogan CH, Owens SM, Reynoso GV, Liao Y, Meyer TJ, Zelazowska MA, Liu B, Li X, Grosskopf AK, Khairallah C, Kirillov V, Reich NC, Sheridan BS, McBride KM, Gewurz BE, Hickman HD, Forrest JC, Krug LT. mBio. 2024 Feb 14;15(2):e0299823.
Kuang X, Salinger A, Benavides F, Muller WJ, Dent SYR*, Koutelou E*. PLoS One. 2024 Jan 18;19(1):e0290837. (*Co-corresponding authors)
Larouche JD, Laumont CM, Trofimov A, Vincent K, Hesnard L, Brochu S, C?t¨¦ C, Humeau JF, Bonneil ?, Lanoix J, Durette C, Gendron P, Laverdure JP, Richie ER, Lemieux S, Thibault P, Perreault C. Elife. 2024 Apr 18;12:RP91037.
Lee J, Chatterjee B, Oh N, Saha D, Lu Y, Bartholomew B, Ishak CA. J Vis Exp. 2024 Dec 13;(214). doi: 10.3791/67359.PMID: 39760380
Malone CF, Mabe NW, Forman AB, Alexe G, Engel KL, Chen YC, Soeung M, Salhotra S, Basanthakumar A, Liu B, Dent SYR, Stegmaier K. Sci Adv. 2024 May 31;10(22):eadm9449.
Marshall VA, Cornejo Castro EM, Goodman CA, Labo N, Liu I, Fisher NC, Moore KN, Nair A, Immonen T, Keele BF, Polizzotto MN, Uldrick TS, Mu Y, Saswat T, Krug LT, McBride KM, Lurain K, Ramaswami R, Yarchoan R, Whitby D. PLoS Pathog. 2024 Jul 15;20(7):e1012338.
Mise K, Long J, Galvan DL, Ye Z, Fan G, Sharma R, Serysheva II, Moore TI, Jeter CR, Anna Zal M, Araki M, Wada J, Schumacker PT, Chang BH, Danesh FR. Nat Commun. 2024 Mar 4;15(1):1965.
Patel B, Zhou Y, Babcock RL, Ma F, Zal MA, Kumar D, Medik YB, Kahn LM, Pineda JE, Park EM, Schneider SM, Tang X, Raso MG, Jeter CR, Zal T, Clise-Dwyer K, Keyomarsi K, Giancotti FG, Colla S, Watowich SS.. Leukemia. 2024 May;38(5):1143-1155.
Pourebrahim R, Montoya RH, Akiyama H, Ostermann L, Khazaei S, Muftuoglu M, Baran N, Zhao R, Lesluyes T, Liu B, Khoury JD, Gagea M, Van Loo P, Andreeff M. Cell Rep Med. 2024 May 21;5(5):101558.
Saha D, Animireddy S, Bartholomew B. Biochem Soc Trans. 2024 Apr 24;52(2):603-616.
Saha D, Animireddy S, Lee J, Thommen A, Murvin MM, Lu Y, Calabrese JM, Bartholomew B. Mol Cell. 2024 May 16;84(10):1855-1869.e5.
Sarkar S, Stitzlein LM, Chandra J. Pediatr Med. 2024 Feb 28;7:3. doi: 10.21037/pm-23-43.
Shanley M, Daher M, Dou J, Li S, Basar R, Rafei H, Dede M, Gumin J, Pantale?n Garc?a J, Nunez Cortes AK, He S, Jones CM, Acharya S, Fowlkes NW, Xiong D, Singh S, Shaim H, Hicks SC, Liu B, Jain A, Zaman MF, Miao Q, Li Y, Uprety N, Liu E, Muniz-Feliciano L, Deyter GM, Mohanty V, Zhang P, Evans SE, Shpall EJ, Lang FF, Chen K, Rezvani K. Cancer Cell. 2024 Aug 12;42(8):1450-1466.e11.
Sharma M, Barravecchia I, Teis R, Cruz J, Mumby R, Ziemke EK, Espinoza CE, Krishnamoorthy V, Magnuson B, Ljungman M, Koschmann C, Chandra J, Whitehead CE, Sebolt-Leopold JS, Galban S. Mol Cancer Ther. 2024 Jan 3;23(1):24-34.
Shen L, Ma X, Wang Y, Wang Z, Zhang Y, Pham HQH, Tao X, Cui Y, Wei J, Lin D, Abeywanada T, Hardikar S, Halabelian L, Smith N, Chen T, Barsyte-Lovejoy D, Qiu S, Xing Y, Yang Y. Nat Commun. 2024 Apr 1;15(1):2809.
Shukla S, Paul S, Garcia J, Zhong Y, Juste SS, Beauchemin K, Bartholomew B. Nucleic Acids Res. 2024 Dec 16:gkae1187. doi: 10.1093/nar/gkae1187.
Stitzlein LM, Adams JT, Stitzlein EN, Dudley RW, Chandra J. J Exp Clin Cancer Res. 2024 Jan 5;43(1):12.
Teboul L, Amos-Landgraf J, Benavides FJ, Birling MC, Brown SDM, Bryda E, Bunton-Stasyshyn R, Chin HJ, Crispo M, Delerue F, Dobbie M, Franklin CL, Fuchtbauer EM, Gao X, Golzio C, Haffner R, H¨¦rault Y, Hrabe de Angelis M, Lloyd KCK, Magnuson TR, Montoliu L, Murray SA, Nam KH, Nutter LMJ, Pailhoux E, Pardo Manuel de Villena F, Peterson K, Reinholdt L, Sedlacek R, Seong JK, Shiroishi T, Smith C, Takeo T, Tinsley L, Vilotte JL, Warming S, Wells S, Whitelaw CB, Yoshiki A; Asian Mouse Mutagenesis Resource Association; CELPHEDIA infrastructure; INFRAFRONTIER consortium; International Mammalian Genome Society; International Mouse Phenotyping Consortium; International Society for Transgenic Technologies; Mutant Mouse Resource and Research Centers; Phenomics Australia; RRRC- Rat Resource and Research Center; Pavlovic G. Nat Commun. 2024 Jul 2;15(1):5574.
Tiniakou I, Hsu PF, Lopez-Zepeda LS, Garipler G, Esteva E, Adams NM, Jang G, Soni C, Lau CM, Liu F, Khodadadi-Jamayran A, Rodrick TC, Jones D, Tsirigos A, Ohler U, Bedford MT, Nimer SD, Kaartinen V, Mazzoni EO, Reizis B. Sci Immunol. 2024 Apr 12;9(94):eadi1023.
Tzetzo SL, Kramer ED, Mohammadpour H, Kim M, Rosario SR, Yu H, Dolan MR, Oturkar CC, Morreale BG, Bogner PN, Stablewski AB, Benavides FJ, Brackett CM, Ebos JML, Das GM, Opyrchal M, Nemeth MJ, Evans SS, Abrams SI. iScience. 2024 Feb 10;27(3):109187.
Wible DJ, Parikh Z, Cho EJ, Chen MD, Mukhopadhyay S, Dalby KN, Varadarajan S, Bratton SB. . Cell Death Dis. 2024 Jan 22;15(1).
Viner C, Ishak CA, Johnson J, Walker NJ, Shi H, Sj?berg-Herrera MK, Shen SY, Lardo SM, Adams DJ, Ferguson-Smith AC, De Carvalho DD, Hainer SJ, Bailey TL, Hoffman MM. Genome Biol. 2024 Jan 8;25(1):11.
Xuan F, Xuan H, Huang M, He W, Xu H, Shi X, Wen H. J Mol Biol. 2024 Apr 1;436(7):168414.
Zhao F, Deng Y, Yang F, Yan Y, Feng F, Peng B, Gao J, Bedford MT, Li H. J Mol Biol. 2024 Apr 1;436(7):168371.
Zhang X, Blumenthal RM, Cheng X. Epigenomes. 2024 Jun 19;8(2):23.
Zhang X, Blumenthal RM, Cheng X. Curr Opin Struct Biol. 2024 Aug;87:102836.
Zhang X, Xia F, Zhang X, Blumenthal RM, Cheng X. J Mol Biol. 2024 Apr 1;436(7):168343.
Zehrbach NM, Oh N, Ishak CA. Trends Cancer. 2024 Apr;10(4):286-288.
Zhou J, Chen Q, Ren R, Yang J, Liu B, Horton JR, Chang C, Li C, Maksoud L, Yang Y, Rotili D, Jain AK, Zhang X, Blumenthal RM, Chen T, Gao Y, Valente S, Mai A, Cheng X. Cell Chem Biol. 2024 Dec 19;31(12):2112-2127.e6.
2023
Beilinson HA, Sevilleja A, Spring J, Benavides F, Beilinson V, Neokosmidis N, Golovkina T. J Virol. 2023 Aug 31;97(8):e0068523.
Chen Q, Liu B, Zeng Y, Hwang JW, Dai N, Corr¨ºa IR Jr, Estecio MR, Zhang X, Santos MA, Chen T, Cheng X. NAR Cancer. 2023 May 17;5(2):zcad022.
Dan J, Du Z, Zhang J, Chen T. Cancer Biol Med. 2023 Aug 18;20(8):545-52.
Fan H, Wang F, Zeng A, Murison A, Tomczak K, Hao D, Jelloul FZ, Wang B, Barrodia P, Liang S, Chen K, Wang L, Zhao Z, Rai K, Jain AK, Dick J, Daver N, Futreal A, Abbas HA. Commun Biol. 2023 Jul 21;6(1):765.
Figueroa M, Ma H, Alfayez M, Morales-Mantilla DE, Wang F, Lu Y, Estecio M, King KY, Kleinerman ES, Moghaddam SJ, Daver NG, Andreeff M, Konopleva MY, DiNardo CD, Chandra J. Blood Adv. 2023 Nov 14;7(21):6624-6629.
Gao G, Hausmann S, Flores NM, Benitez AM, Shen J, Yang X, Person MD, Gayatri S, Cheng D, Lu Y, Liu B, Mazur PK, Bedford MT. Nat Commun. 2023 Jan 23;14(1):363.
Gencel-Augusto J, Su X, Qi Y, Whitley EM, Pant V, Xiong S, Shah V, Lin J, Perez E, Fiorotto ML, Mahmud I, Jain AK, Lorenzi PL, Navin NE, Richie ER, Lozano G. Cancer Discov. 2023 May 4;13(5):1230-1249.
Horton JR, Zhou J, Chen Q, Zhang X, Bedford MT, Cheng X. J Biol Chem. 2023 Feb;299(2):102862.
Hussain T, Sanchez K, Crayton J, Saha D, Jeter C, Lu Y, Abba M, Seo R, Noebels JL, Fonken L, Aldaz CM. Prog Neurobiol. 2023 Apr;223:102425.
Liu KJ, Zelazowska MA, McBride KM. Viruses. 2023 May 26;15(6):1253.
Mahadevan KK, LeBleu VS, Ramirez EV, Chen Y, Li B, Sockwell AM, Gagea M, Sugimoto H, Sthanam LK, Tampe D, Zeisberg M, Ying H, Jain AK, DePinho RA, Maitra A, McAndrews KM, Kalluri R. Dev Cell. 2023 May 4;13(5):1230-1249.
Mu Y, Zelazowska MA, Chen Z, Plummer JB, Dong Q, Krug LT, McBride KM. DNA Repair (Amst). 2023 Aug;128:103515.
Patel LR, Stratton SA, McLaughlin M, Krause P, Allton K, Rivas AL, Barbosa D, Hart T, Barton MC. iScience. 2023 Jun 14;26(7):107126.
Premkumar T, Paniker L, Kang R, Biot M, Humphrey E, Destain H, Ferranti I, Okulate I, Nguyen H, Kilaru V, Frasca M, Chakraborty P, Cole F. Mol Cell. 2023 Aug 17;83(16):2941-2958.e7.
Saha D, Hailu S, Hada A, Lee J, Luo J, Ranish JA, Lin YC, Feola K, Persinger J, Jain A, Liu B, Lu Y, Sen P, Bartholomew B. Nat Commun. 2023 Aug 4;14(1):4682.
Santos M, Hwang JW, Bedford MT. J Biol Chem. 2023 Aug 1:105124.
Smith KR, Paul S, Dong Q, Anannya O, Oldenburg DG, Forrest JC, McBride KM, Krug LT. mSphere. 2023 Oct 24;8(5):e0027823.
Srinivasan J, Vasudev A, Shasha C, Selden HJ, Perez E Jr, LaFleur B, Sinari SA, Krueger A, Richie ER, Ehrlich LIR. Aging Cell. 2023 Aug;22(8):e13870.
Stitzlein LM, Gangadharan A, Walsh LM, Nam D, Espejo AB, Singh MM, Patel KH, Lu Y, Su X, Ezhilarasan R, Gumin J, Singh S, Sulman E, Lang FF, Chandra J. Front Neurol. 2023 Apr 4;14:1112207.
Stroik S, Carvajal-Garcia J, Gupta D, Edwards A, Luthman A, Wyatt DW, Dannenberg RL, Feng W, Kunkel TA, Gupta GP, Hedglin M, Wood R, Doubli¨¦ S, Rothenberg E, Ramsden DA. Nature. 2023 Nov;623(7988):836-841.
Thiyagarajan T, Ponnusamy S, Hwang DJ, He Y, Asemota S, Young KL, Johnson DL, Bocharova V, Zhou W, Jain AK, Petricoin EF, Yin Z, Pfeffer LM, Miller DD, Narayanan R. Proc Natl Acad Sci U S A. 2023 Jan 3;120(1):e2211832120.
Wang Y, Bedford MT. Biochem Soc Trans. 2023 Apr 26;51(2):725-734.
Yang J, Horton JR, Liu B, Corces VG, Blumenthal RM, Zhang X, Cheng X. Nucleic Acids Res. 2023 Sep 8;51(16):8447-8462.
Ying Z, Hardikar S, Plummer JB, Hamidi T, Liu B, Chen Y, Shen J, Mu Y, McBride KM, Chen T. Cell Mol Immunol. 2023 Dec;20(12):1487-1498
Xu L, Xuan H, He W, Zhang L, Huang M, Li K, Wen H, Xu H, Shi X. Nat Commun. 2023 Sep 2;14(1):5362.
Zhang L, He W, Fu R, Wang S, Chen Y, Xu H. Nucleic Acids Res. 2023 Oct 13;51(18):9880-9893.
Zheng C, Wei Y, Zhang P, Xu L, Zhang Z, Lin K, Hou J, Lv X, Ding Y, Chiu Y, Jain A, Islam N, Malovannaya A, Wu Y, Ding F, Xu H, Sun M, Chen X, Chen Y. J Clin Invest. 2023 Mar 1;133(5):e159940.
Zheng C, Wei Y, Zhang P, Lin K, He D, Teng H, Manyam G, Zhang Z, Liu W, Lee HRL, Tang X, He W, Islam N, Jain A, Chiu Y, Cao S, Diao Y, Meyer-Gauen S, H??k M, Malovannaya A, Li W, Hu M, Wang W, Xu H, Kopetz S, Chen Y. Nat Struct Mol Biol. 2023 Nov 6. Online ahead of print
Zhou J, Deng Y, Iyamu ID, Horton JR, Yu D, Hajian T, Vedadi M, Rotili D, Mai A, Blumenthal RM, Zhang X, Huang R, Cheng X. ACS Chem Biol. 2023 Apr 21;18(4):734-745.
Zhou J, Horton JR, Menna M, Fiorentino F, Ren R, Yu D, Hajian T, Vedadi M, Mazzoccanti G, Ciogli A, Weinhold E, H¨¹ben M, Blumenthal RM, Zhang X, Mai A, Rotili D, Cheng X. J Med Chem. 2023 Jan 12;66(1):934-950.
2022
Basu S, Dong Y, Kumar R, Jeter C, Tang DG. Semin Cancer Biol. 2022 Jan;78:90-103.
Carvajal-Maldonado D, Drogalis Beckham L, Wood RD, Doubli¨¦ S. . Front Mol Biosci. 2022 Jan 7;8:815845
Chen YC, Koutelou E, Dent SYR. Mol Cell. 2022 Feb 17;82(4):716-727.
Cheng X, Blumenthal RM. . Curr Opin Struct Biol. 2022 Aug;75:102433.
Dan J, Chen T. . Adv Exp Med Biol. 2022;1389:111-136.
Darbaniyan F, Zheng H, Kanagal-Shamanna R, Lockyer P, Montalban-Bravo G, Estecio M, Lu Y, Soltysiak KA, Chien KS, Yang H, Sasaki K, Class C, Ganan-Gomez I, Do KA, Garcia-Manero G, Wei Y. . Exp Hematol. 2022 Nov;115:44-53.
Fabre ML, Canzoneri R, Gurruchaga A, Lee J, Tatineni P, Kil H, Lacunza E, Aldaz CM*, Abba MC*. . Cancers (Basel). 2022 Jun 7;14(12):2819. (*co-corresponding authors)
Fedoriw A#, Shi L#, O'Brien S, Smitheman KN, Wang Y, Hou J, Sherk C, Rajapurkar S, Laraio J, Williams LJ, Xu C, Han G, Feng Q, Bedford MT, Wang L, Barbash O, Kruger RG, Hwu P, Mohammad HP, Peng W. . Cancer Immunol Res. 2022 Apr 1;10(4):420-436. (#equal contribution)
Fontes F, Rocha S, S¨¢nchez R, Pessina P, Sebastian M, Benavides F, Breijo M. . Lab Anim. 2022 Mar 31:236772221085356. doi: 10.1177/00236772221085356.
Fu* R, He W*, Dou J, Villarreal OD, Bedford E, Wang H, Hou C, Zhang L, Wang Y, Ma D, Chen Y, Gao X, Depken M, Xu H. . Nat Commun. 2022 Jan 25;13(1):474. (*equal contribution)
Horton JR, Pathuri S, Wong K, Ren R, Rueda L, Fosbenner DT, Heerding DA, McCabe MT, Pappalardi MB, Zhang X, King BW, Cheng X. . Structure. 2022 Jun 2;30(6):793-802.e5.
Huang P, Peslak SA, Ren R, Khandros E, Qin K, Keller CA, Giardine B, Bell HW, Lan X, Sharma M, Horton JR, Abdulmalik O, Chou ST, Shi J, Crossley M, Hardison RC, Cheng X, Blobel GA. . Nat Genet. 2022 Sep;54(9):1417-1426
Huang YH, Chen CW, Sundaramurthy V, S?abicki M, Hao D, Watson CJ, Tovy A, Reyes JM, Dakhova O, Crovetti BR, Galonska C, Lee M, Brunetti L, Zhou Y, Tatton-Brown K, Huang Y, Cheng X, Meissner A, Valk PJM, Van Maldergem L, Sanders MA, Blundell JR, Li W, Ebert BL, Goodell MA. Cancer Discov. 2022 Jan;12(1):220-235.
Iannelli G, Milite C, Marechal N, Cura V, Bonnefond L, Troffer-Charlier N, Feoli A, Rescigno D, Wang Y, Cipriano A, Viviano M, Bedford MT, Cavarelli J, Castellano S, Sbardella G. . J Med Chem. 2022 Apr 28. doi: 10.1021/acs.jmedchem.2c00252.
Kannan S, Irwin ME, Herbrich SM, Cheng T, Patterson LL, Aitken MJL, Bhalla K, You MJ, Konopleva M, Zweidler-McKay PA, Chandra J. . Antioxidants (Basel). 2022 Apr 5;11(4):717.
Khan FI, Rehman T, Sameena F, Hussain T*, AlAjmi MF, Lai D, Khan MKA. . J Mol Recognit. 2022 Jul;35(7):e2958. (*postdoctoral fellow with C.M. Aldaz)
Kim KB, Kabra A, Kim DW, Xue Y*, Huang Y, Hou PC, Zhou Y, Miranda LJ, Park JI, Shi X, Bender TP, Bushweller JH, Park KS. . Sci Adv. 2022 Feb 18;8(7):eabl4618. (*graduate student with X. Shi)
Koutelou E, Dent SYR. . Nat Cell Biol. 2022 Apr;24(4):412-414. (News & Views article)
Lancaster JN, Keatinge-Clay DE, Srinivasan J, Li Y, Selden HJ, Nam S, Richie ER, Ehrlich LIR. . Aging Cell. 2022 Jun;21(6):e13624. PMID: 35561351
Liao Y, Chen CH, Xiao T, de la Pe?a Avalos B, Dray EV, Cai C, Gao S, Shah N, Zhang Z, Feit A, Xue P, Liu Z, Yang M, Lee JH, Xu H, Li W, Mei S, Pierre RS, Shu S, Fei T, Duarte M, Zhao J, Bradner JE, Polyak K, Kantoff PW, Long H, Balk SP, Liu XS, Brown M, Xu K. . Proc Natl Acad Sci U S A. 2022 Jan 18;119(3):e2105898119.
Liu Y, Iqbal A, Li W, Ni Z, Wang Y, Ramprasad J, Abraham KJ, Zhang M, Zhao DY, Qin S, Loppnau P, Jiang H, Guo X, Brown PJ, Zhen X, Xu G, Mekhail K, Ji X, Bedford MT, Greenblatt JF, Min J. . Nat Commun. 2022 Sep 16;13(1):5453.
Meng H, Gonzales NM, Jung SY, Lu Y, Putluri N, Zhu B, Dacso CC, Lonard DM, O'Malley BW. . Cell Rep. 2022 Mar 8;38(10):110491.
Miglioranza Scavuzzi B, van Drongelen V, Kaur B, Fox JC, Liu J, Mesquita-Ferrari RA, Kahlenberg JM, Farkash EA, Benavides F, Miller FW, Sawalha AH, Holoshitz J. . Commun Biol. 2022 Jul 28;5(1):751.
Moyret-Lalle C, Prodhomme MK*, Burlet D, Kashiwagi A, Petrilli V, Puisieux A, Seimiya H, Tissier A. Role of EMT in the DNA damage response, double-strand break repair pathway choice and its implications in cancer treatment. Cancer Sci. 2022 May 9. doi: 10.1111/cas.15389. (*Prodhomme is a postdoc in the Wood lab)
Mustachio LM, Roszik J . . Cancers (Basel). 2022 Jan 28;14(3):657.
Ozirmak Lermi N, Gray SB, Bowen CM, Reyes-Uribe L, Dray BK, Deng N, Harris RA, Raveendran M, Benavides F, Hodo CL, Taggart MW, Colbert Maresso K, Sinha KM, Rogers J, Vilar E. PLoS Genet. 2022 Apr 21;18(4):e1010163.
Park D, Gharghabi M, Reczek CR, Plow R, Yungvirt C, Aldaz CM, Huebner K. . Int J Mol Sci. 2022 Mar 28;23(7):3729.
Perez CJ, Mecklenburg L, Fernandez A, Cantero M, de Souza T, Lin K, Dent SYR, Montoliu L, Awgulewitsch A, Benavides, F. 2022 Mar;31(3):330-340.
Piya S, Yang Y, Bhattacharya S, Sharma P, Ma H, Mu H, He H, Ruvolo V, Baran N, Davis RE, Jain AK, Konopleava M, Kantarjian H, Andreeff M, You MJ, Borthakur G. . Leukemia. 2022 May;36(5):1261-1273.
Ren R#, Horton JR#, Hong S, Cheng X. . Adv Exp Med Biol. 2022;1389:295-315. (#equal contribution)
Soundararajan R, Viscuse P, Pilie P, Liu J, Logotheti S, Laberiano Fern¨¢ndez C, Lorenzini D, Hoang A, Lu W, Soto LMS, Wistuba II, Xu M, Song X, Shepherd PDA, Navone NM, Tidwell RSS, Lozano G, Logothetis C, Zhang J, Long JP, Estecio MR, Tzelepi V, Aparicio AM. . Cancers (Basel). 2022 Jun 30;14(13):3233.
Srour N, Villarreal OD, Hardikar S, Yu Z, Preston S, Miller WH Jr, Szewczyk MM, Barsyte-Lovejoy D, Xu H, Chen T, del Rinc¨®n SV, Richard S. . Cell Rep. 2022 Mar 29;38(13):110582.
Tu SM, Estecio MR, Lin SH, Zacharias NM. Cancers (Basel). 2022 Jan 27;14(3):655.
Van HT, Harkins PR, Patel A, Jain AK, Lu Y, Bedford MT, Santos MA. . J Biol Chem. 2022 Mar;298(3):101588.
Vanson S, Li Y, Wood RD, Doubli¨¦ S. DNA Repair (Amst). 2022 Aug;116:103358.
Wang Y, Person MD, Bedford MT. s. Methods. 2022 Apr;200:80-86.
Wang Z, Lu Y, Fornage M, Jiao L, Shen J, Li D, Wei P. . Epigenetics. 2022 Nov;17(11):1357-1372.
Wood RD, Doubli¨¦ S. Annu Rev Genet. 2022 Nov 30;56:207-228.
Yang J*, Gupta E*, Horton JR, Blumenthal RM, Zhang X, Cheng X. DNA Repair. . DNA Repair DNA Repair (Amst). 2022 Apr;298(4):101751. (*equal contribution)
Yu D, Dai N, Wolf EJ, Corr¨ºa IR Jr, Zhou J, Wu T, Blumenthal RM, Zhang X, Cheng X. J Biol Chem. 2022 Apr;298(4):101751
Yu D, Zhou J, Chen Q, Wu T, Blumenthal RM, Zhang X, Cheng X. . Biochemistry. 2022 May 23;61(11):1005-13.
Zhou J, Horton JR, Yu D, Ren R, Blumenthal RM, Zhang X, Cheng X. . Epigenetics. 2022 Sep;17(9):970-981.
Zhao S, Habib SL, Senejani AG, Sebastian M, Kidane D. . Biomedicines. 2022 Feb 26;10(3):557.
Zhang Y, Li Y, Chachad D, Liu B, Godavarthi JD, Williams-Villalobo A, Lasisi L, Xiong S, Matin A. Biochem Biophys Rep. 2022 Jan 13;29:101206.
Zhang Y, Williams-Villalobo A, Godavarthi JD, Shakoor F, Xiong S, Liu B. Biochem Biophys Rep. 2022 Dec 7;33:101404.
2021
Abba MC, Fabre ML, Lee J, Tatineni P, Kil H, Aldaz CM. . Front Oncol. 2021 Nov 17;11:783211.
Bailey CP, Wang R, Figueroa M, Zhang S, Wang L, Chandra J. . Comput Syst Oncol. 2021 Sep;1(3):e1016.
Ben Yamin B#, Ahmed-Seghir S#, Tomida J#, Despras E, Pouvelle C, Yurchenko A, Goulas J, Corre R, Delacour Q, Droin N, Dessen P, Goidin D, Lange SS, Bhetawal S, Mitjavila-Garcia MT, Baldacci G, Nikolaev S, Cadoret JC*, Wood RD*, Kannouche PL*. . EMBO J. 2021 Nov 2;40(21):e104543. (#equal contributions as first authors, *equal contributions as senior authors)
Carvajal-Maldonado D, Wood RD. . Cancer Res. 2021 Mar 15;81(6):1441-1442.
Chen J, Horton J, Sagum C, Zhou J, Cheng X, Bedford MT. Biochem J. 2021 May 28;478(10):1943-1958.
Chen LM, Chai JC, Liu B, Strutt TM, McKinstry KK, Chai KX. . Biosci Rep. 2021 Jul 30;41(7):BSR20211370.
Cheng T, Kiser K, Grasse L, Iles L, Bartholomeusz G, Samaniego F, Orlowski RZ, Chandra J. . Cancer Drug Resist. 2021;4(4):888-902.
Chen YC, Dent SYR. . Epigenetics Chromatin. 2021 Jun 10;14(1):26.
Dejima H, Hu X, Chen R, Zhang J, Fujimoto J, Parra ER, Haymaker C, Hubert SM, Duose D, Solis LM, Su D, Fukuoka J, Tabata K, Pham HHN, Mcgranahan N, Zhang B, Ye J, Ying L, Little L, Gumbs C, Chow CW, Estecio MR, Godoy MCB, Antonoff MB, Sepesi B, Pass HI, Behrens C, Zhang J, Vaporciyan AA, Heymach JV, Scheet P, Lee JJ, Wu J, Futreal PA, Reuben A, Kadara H, Wistuba II, Zhang J. . Nat Commun. 2021 May 11;12(1):2722.
DiNardo CD, Beird HC, Estecio M, Hardikar S, Takahashi K, Bannon SA, Borthakur G, Jabbour E, Gumbs C, Khoury JD, Routbort M, Gong T, Kondo K, Kantarjian H, Garcia-Manero G, Chen T, Futreal PA. . Epigenetics. 2021 May;16(5):567-576.
Genois MM, Gagn¨¦ JP, Yasuhara T, Jackson J, Saxena S, Langelier MF, Ahel I, Bedford MT, Pascal JM, Vindigni A, Poirier GG, Zou L. . Mol Cell. 2021 Feb 18;81(4):784-800.e8
He W, Wang H, Wei Y, Jiang Z, Tang Y, Chen Y, Xu H. . Bioinformatics. 2021 Jan 4:btaa1068.
Hu X, Estecio MR, Chen R, Reuben A, Wang L, Fujimoto J, Carrot-Zhang J, McGranahan N, Ying L, Fukuoka J, Chow CW, Pham HHN, Godoy MCB, Carter BW, Behrens C, Zhang J, Antonoff MB, Sepesi B, Lu Y, Pass HI, Kadara H, Scheet P, Vaporciyan AA, Heymach JV, Wistuba II, Lee JJ, Futreal PA, Su D, Issa JJ, Zhang J. . Nat Commun. 2021 Jan 29;12(1):687.
Jung YS, Stratton SA, Lee SH, Kim MJ, Jun S, Zhang J, Zheng B, Cervantes CL, Cha JH, Barton MC, Park JI. TMEM9-v-ATPase Activates Wnt/¦Â-Catenin Signaling via APC Lysosomal Degradation for Liver Regeneration and Tumorigenesis. Hepatology. 2021 Feb;73(2):776-794.
Koutelou E, Farria AT, Dent SYR. . Biochim Biophys Acta Gene Regul Mech. 2021 Feb;1864(2):194609.
Kuang X, McAndrew MJ, Mustachio LM, Chen YC, Atanassov BS, Lin K, Lu Y, Shen J, Salinger A, Macatee T, Dent SYR*, Koutelou E*. Cancers (Basel). 2021 Aug 25;13(17):4276. (*co-corresponding authors)
Lan X, Ren R, Feng R, Ly LC, Lan Y, Zhang Z, Aboreden N, Qin K, Horton JR, Grevet JD, Mayuranathan T, Abdulmalik O, Keller CA, Giardine B, Hardison RC, Crossley M, Weiss MJ, Cheng X, Shi J, Blobel GA. . Mol Cell. 2021 Jan 21;81(2):239-254.e8.
Llorens-Agost M, Ensminger M, Le HP, Gawai A, Liu J, Cruz-Garc¨ªa A, Bhetawal S, Wood RD, Heyer WD, L?brich M. . Nat Cell Biol. 2021 Oct;23(10):1095-1104.
Manickavinayaham S, Dennehey BK, Johnson DG. Cancers (Basel). 2021 Feb 24;13(5):934.
Martin SK, Tomida J, Wood RD. . Cell Rep. 2021 Feb 23;34(8):108775.
Morales F, Corrales E, Zhang B, V¨¢squez M, Santamar¨ªa-Ulloa C, Quesada H, Sirito M, Estecio MR, Monckton DG, Krahe R. . Hum Mol Genet. 2021 Dec 27;31(2):262-274.
Muradova E, Patel N, Sell B, Bittencourt BB, Ojeda SS, Adelmann CH, Cen L, Cheng CH, Shen J, Davis CM, Ehli EA, Newberg JY, Cherpelis B, Black MA, Mann MB, Mitragotri S, Tsai KY. . J Invest Dermatol. 2021 Jan;141(1):124-131.e2.
Pappalardi MB, Keenan K, Cockerill M, Kellner WA, Stowell A, Sherk C, Wong K, Pathuri S, Briand J, Steidel M, Chapman P, Groy A, Wiseman AK, McHugh CF, Campobasso N, Graves AP, Fairweather E, Werner T, Raoof A, Butlin RJ, Rueda L, Horton JR, Fosbenner DT, Zhang C, Handler JL, Muliaditan M, Mebrahtu M, Jaworski JP, McNulty DE, Burt C, Eberl HC, Taylor AN, Ho T, Merrihew S, Foley SW, Rutkowska A, Li M, Romeril SP, Goldberg K, Zhang X, Kershaw CS, Bantscheff M, Jurewicz AJ, Minthorn E, Grandi P, Patel M, Benowitz AB, Mohammad HP, Gilmartin AG, Prinjha RK, Ogilvie D, Carpenter C, Heerding D, Baylin SB, Jones PA, Cheng X, King BW, Luengo JI, Jordan AM, Waddell I, Kruger RG, McCabe MT. . Nat Cancer. 2021 Oct;2(10):1002-1017.
Park D, Gharghabi M, Schrock MS, Plow R, Druck T, Yungvirt C, Aldaz CM, Huebner K. . DNA Repair (Amst). 2022 Feb;110:103264.
Shah VV*, Duncan AD*, Jiang S*, Stratton SA, Allton KL, Yam C, Jain A, Krause PM, Lu Y, Cai S, Tu Y, Zhou X, Zhang X, Jiang Y, Carroll CL, Kang Z, Liu B, Shen J, Gagea M, Manu SM, Huo L, Gilcrease M, Powell RT, Guo L, Stephan C, Davies PJ, Parker-Thornburg J, Lozano G, Behringer RR, Piwnica-Worms H, Chang JT, Moulder SL, Barton MC. . Nat Commun. 2021 Sep 10;12(1):5389. (*co-first authors)
Srinivasan J, Lancaster JN, Singarapu N, Hale LP, Ehrlich LIR, Richie ER. Front Immunol. 2021 Apr 22;12:676236. (Review)
Sun M#, Wang Y#, Zheng C#, Wei Y, Hou J, Zhang P, He W, Lv X, Ding Y, Liang H, Hon CC, Chen X, Xu H*, Chen Y*. . Genome Biol. 2021 Aug 23;22(1):240. (#equal contribution, *co-corresponding authors)
Toraason E, Horacek A, Clark C, Glover ML, Adler VL, Premkumar T, Salagean A, Cole F, Libuda DE. . Curr Biol. 2021 Apr 12;31(7):1508-1514.e5.
Trinh A, Gil Del Alcazar CR, Shukla SA, Chin K, Chang YH, Thibault G, Eng J, Jovanovi? B, Aldaz CM, Park SY, Jeong J, Wu C, Gray J, Polyak K. . Mol Cancer Res. 2021 Apr;19(4):623-635.
Wright T, Wang Y, Bedford MT. . Epigenomes. 2021 Mar;5(1):2. (Review)
Yang F, Chen J, Liu B, Gao G, Sebastian M, Jeter C, Shen J, Person MD, Bedford MT. . Nat Commun. 2021 Nov 4;12(1):6362.
Yang J, Horton JR, Akdemir KC, Li J, Huang Y, Kumar J, Blumenthal RM, Zhang X, Cheng X. Nucleic Acids Res. 2021 May 21;49(9):5084-5094.
Yang Y, Ren R, Ly LC, Horton JR, Li F, Quinlan KGR, Crossley M, Shi Y, Cheng X. Cell Rep. 2021 Sep 28;36(13):109759.
Yu D#, Horton JR#, Yang J, Hajian T, Vedadi M, Sagum CA, Bedford MT, Blumenthal RM, Zhang X, Cheng X. . Nucleic Acids Res. 2021 Nov 18;49(20):11629-11642. (#equal contribution)
Zhou J#, Horton JR#, Blumenthal RM, Zhang X, Cheng X. Nat Commun. 2021 Jun 8;12(1):3436. (#equal contribution)
2020
Abba MC, Canzoneri R, Gurruchaga A, Lee J, Tatineni P, Kil H, Lacunza E, Aldaz CM. . Int J Mol Sci. 2020 Oct 8;21(19):7407.
Benavides F, R¨¹licke T, Prins JB, Bussell J, Scavizzi F, Cinelli P, Herault Y, Wedekind D. . Lab Anim. 2020 Apr;54(2):135-148.
Bhardwaj SK, Hailu SG, Olufemi L, Brahma S, Kundu S, Hota SK, Persinger J, Bartholomew B. . Nat Commun. 2020 Nov 20;11(1):5913.
Farria AT, Plummer JB, Salinger AP, Shen J, Lin K, Lu Y, McBride KM, Koutelou E, Dent SYR. . Cancer Res. 2020 Dec 15;80(24):5543-5553.
Hardikar S, Ying Z, Zeng Y, Zhao H, Liu B, Veland N, McBride K, Cheng X, Chen T. . Protein Cell. 2020 Mar;11(3):214-218.
Helmink BA, Reddy SM, Gao J, Zhang S, Basar R, Thakur R, Yizhak K, Sade-Feldman M, Blando J, Han G, Gopalakrishnan V, Xi Y, Zhao H, Amaria RN, Tawbi HA, Cogdill AP, Liu W, LeBleu VS, Kugeratski FG, Patel S, Davies MA, Hwu P, Lee JE, Gershenwald JE, Lucci A, Arora R, Woodman S, Keung EZ, Gaudreau PO, Reuben A, Spencer CN, Burton EM, Haydu LE, Lazar AJ, Zapassodi R, Hudgens CW, Ledesma DA, Ong S, Bailey M, Warren S, Rao D, Krijgsman O, Rozeman EA, Peeper D, Blank CU, Schumacher TN, Butterfield LH, Zelazowska MA, McBride KM, Kalluri R, Allison J, Petitprez F, Fridman WH, Saut¨¨s-Fridman C, Hacohen N, Rezvani K, Sharma P, Tetzlaff MT, Wang L, Wargo JA. . Nature. 2020 Jan;577(7791):549-555.
Hwang T, Reh S, Dunbayev Y, Zhong Y, Takata Y, Shen J, McBride KM, Murnane JP, Bhak J, Lee S, Wood RD, Takata KI. . NAR Cancer. 2020 Sep;2(3):zcaa017.
Jain K, Fraser CS, Marunde MR, Parker MM, Sagum C, Burg JM, Hall N, Popova IK, Rodriguez KL, Vaidya A, Krajewski K, Keogh MC, Bedford MT*, Strahl BD.* Epigenetics Chromatin. 2020 Jan 24;13(1):3 (*Co-corresponding authors)
Kumar A, Zhong Y, Albrecht A, Sang PB, Maples A, Liu Z, Vinayachandran V, Reja R, Lee CF, Kumar A, Chen J, Xiao J, Park B, Shen J, Liu B, Person MD, Trybus KM, Zhang KYJ, Pugh BF, Kamm KE, Milewicz DM, Shen X*, Kapoor P*. . Cell Rep. 2020 Sep 29;32(13):108172. (*co-corresponding authors)
Kumar J, Kaur G, Ren R, Lu Y, Lin K, Li J, Huang Y, Patel A, Barton MC, Macfarlan T, Zhang X, Cheng X. NAR Cancer. 2020 Jun;2(2):zcaa007.
Shen Y, Gao G, Yu X, Kim H, Wang L, Xie L, Schwarz M, Chen X, Guccione E, Liu J*, Bedford MT*, Jin J*. . J Med Chem. 2020 Sep 10;63(17):9977-9989. (*Co-corresponding authors)
Turner OC, Aeffner F, Bangari DS, High W, Knight B, Forest T, Cossic B, Himmel LE, Rudmann DG, Bawa B, Muthuswamy A, Aina OH, Edmondson EF, Saravanan C, Brown DL, Sing T, Sebastian MM. . Toxicol Pathol. 2020 Feb;48(2):277-294.
Veazey KJ, Cheng D, Lin K, Villarreal OD, Gao G, Perez-Oquendo M, Van HT, Stratton SA, Green M, Xu H, Lu Y, Bedford MT, Santos MA. . Leukemia. 2020 Jun 24. doi: 10.1038/s41375-020-0908-8.
Woodcock CB, Horton JR, Zhou J, Bedford MT, Blumenthal RM, Zhang X, Cheng X. Nucleic Acids Res. 2020 Oct 9;48(18):10329-10341.
Zelazowska MA, Dong Q, Plummer JB, Zhong Y, Liu B, Krug LT, McBride KM. . Life Sci Alliance. 2020 Feb 6;3(3):e201900526.
Zeng Y*, Ren R*, Kaur G, Hardikar S, Ying Z, Babcock L, Gupta E, Zhang X, Chen T#, Cheng X#. . Genes Dev. 2020 Oct 1;34(21-22):1546-58. (*co-first authors, #co-corresponding authors)
2019
Bao J, Di Lorenzo A, Lin K, Lu Y, Zhong Y, Sebastian MM, Muller WJ, Yang Y, Bedford MT. (2019) Cancer Res. 79(1):21-32.
Gao G, Zhang L, Villarreal OD, He W, Su D, Bedford E, Moh P, Shen J, Shi X, Bedford MT, Xu H. (2019) . Nucleic Acids Res. 47(10):5038-5048.
He W, Zhang L, Villarreal OD, Fu R, Bedford E, Dou J, Patel AY, Bedford MT, Shi X, Chen T, Bartholomew B, Xu H. (2019) . Nat Commun. 10(1):4541.
Hussain T, Kil H, Hattiangady B, Lee J, Kodali M, Shuai B, Attaluri S, Takata Y, Shen J, Abba MC, Shetty AK, Aldaz CM. (2019) Neurobiol Dis. 121:163-176.
Koutelou E, Wang L, Schibler A, Chao HP, Kuang X, Lin K, Lu Y, Shen J, Jeter CR, Salinger A, Wilson M, Chen YC, Atanassov BS, Tang DG, Dent SY. (2019) . Development. Feb 22;146(4). pii: dev174037. ]
Manickavinayaham S, V¨¦lez-Cruz R, Biswas AK, Bedford E, Klein BJ, Kutateladze TG, Liu B, Bedford MT, Johnson DG. (2019) Nat Commun. 10(1):4951.
McBride KM, Kil H, Mu Y, Plummer JB, Lee J, Zelazowski MJ, Sebastian M, Abba MC, Aldaz CM. (2019) . Front Oncol. 9:517.
Patel L, Kang R, Rosenberg SC, Qiu Y, Raviram R, Chee S, Hu R, Ren B, Cole F,* Corbett KD*. (2019) . Nat Struct Mol Biol. 26(3):164-174. (*co-corresponding authors)
Ren R, Hardikar S, Horton JR, Lu Y, Zeng Y, Singh AK, Lin K, Coletta LD, Shen J, Lin Kong CS, Hashimoto H, Zhang X, Chen T, Cheng X. (2019) Nucleic Acids Res. 47(16):8388-8398.
Wible DJ, Chao HP, Tang DG, Bratton SB. (2019) Cell Discov. 5:42.
Veland N, Lu Y, Hardikar S, Gaddis S, Zeng Y, Liu B, Estecio MR, Takata Y, Lin K, Tomida MW, Shen J, Saha D, Gowher H, Zhao H, Chen T. (2019) Nucleic Acids Res. 47(1):152-167.
2018
Appikonda S, Thakkar KN, Shah PK, Dent SYR, Andersen JN, Barton MC. (2018) J Biol Chem. 293(19):7476-7485.
Bao J, Perez CJ, Kim J, Zhang H, Murphy CJ, Hamidi T, Jaubert J, Platt CD, Chou J, Deng M, Zhou MH, Huang Y, Gait¨¢n-Pe?as H, Gu¨¦net JL, Lin K, Lu Y, Chen T, Bedford MT, Dent SY, Richburg JH, Est¨¦vez R, Pan HL, Geha RS, Shi Q, Benavides F. (2018) JCI Insight. 3(16):e99767.
Bao J, Rousseaux S, Shen J, Lin K, Lu Y, Bedford MT. . (2018) Nucleic Acids Res. 46(9):4327-4343.
Brahma S, Ngubo M, Paul S, Udugama M, Bartholomew B. (2018) . Nat Commun. 90(1):3309
Cheng D, Vemulapalli V, Lu Y, Shen J, Aoyagi S, Fry CJ, Yang Y, Foulds CE, Stossi F, Trevi?o LS, Mancini MA, O¡¯Malley BW, Walker CL, Boyer TG, Bedford MT. (2018) Life Sci Alliance. 1(5):e201800117.
Das P, Veazey KJ, Van HT, Kaushik S, Lin K, Lu Y, Ishii M, Kikuta J, Ge K, Nussenzweig A, Santos MA. (2018) . Proc Natl Acad Sci U S A. 2018 Oct 23;115(43):E10137-E10146. pii: 201806019.
Franco HL, Nagari A, Malladi V, Li W, Xi Y, Richardson D, Allton KL, Tanaka K, Li J, Murakami S, Keyomarsi K, Bedford MT, Shi X, Li W, Barton MC, Dent SYR, Kraus WL. (2018) . Genome Res. 28(2):159-170.
Hamidi T, Singh AK, Veland N, Vemulapalli V, Chen J, Hardikar S, Bao J, Fry CJ, Yang V, Lee KA, Guo A, Arrowsmith CH, Bedford MT, Chen T. (2018) . J Biol Chem. 293(33):12770-12780.
Jain AK, Barton MC. (2018) . Development. 145(8): dev158360.
Kaushik S, Liu F, Veazey KJ, Gao G, Das P, Neves LF, Lin K, Zhong Y, Lu Y, Giuliani V, Bedford MT, Nimer SD, Santos MA. (2018) Leukemia. 32(2):499-509.
Kang R, Zelazowski MJ, Cole F. (2018) . Mol Cell. 69(5):725-727.
Singarapu N, Ma K, Reeh KAG, Shen J, Lancaster JN, Yi S, Xie H, Orkin SH, Manley NR, Ehrlich LIR, Jiang N, Richie ER. (2018) Sci Rep. 8(1):14335.
Wang L, Koutelou E, Hirsch C, McCarthy R, Schibler A, Lin K, Lu Y, Jeter C, Shen J, Barton MC, Dent SYR. (2018) . Stem Cell Reports. 10(1):287-299
Wible DJ, Bratton SB. (2018) . Curr Opin Toxicol. 7:28-36.
2017
Bae N, Viviano M, Su X, Lv J, Cheng D, Sagum C, Castellano S, Bai X, Johnson C, Khalil MI, Shen J, Chen K, Li H, Sbardella G, Bedford MT. (2017) . Nat Chem Biol. 13(7):750-756.
Brahma S, Udugama MI, Kim J, Hada A, Bhardwaj SK, Hailu SG, Lee TH, Bartholomew B. (2017) . Nat Commun. 8:15616.
Carr SM, Munro S, Sagum CA, Fedorov O, Bedford MT, La Thangue NB. (2017) Cell Death Differ. 24(12):2139-2149.
Dan J, Rousseau P, Hardikar S, Veland N, Wong J, Autexier C, Chen T. (2017) . Cell Rep. 20(8):1936-1949.
Espejo AB, Gao G, Black K, Gayatri S, Veland N, Kim J, Chen T, Sudol M, Walker C, Bedford MT. (2017) . J Biol Chem. 292(6):2255-2265.
Li J, Xi Y, Li W, McCarthy RL, Stratton SA, Zou W, Li W, Dent SY, Jain AK, Barton MC. (2017) . Oncogene. 36(21):2991-3001.
Manandhar M, Lowery MG, Boulware KS, Lin KH, Lu Y, Wood RD. (2017) DNA Repair (Amst). 57:76-90.
Mi W, Guan H, Lyu J, Zhao D, Xi Y, Jiang S, Andrews FH, Wang X, Gagea M, Wen H, Tora L, Dent SYR, Kutateladze TG, Li W, Li H, Shi X. (2017) . Nat Commun. 8(1):1088
Mu Y, Zelazowska MA, McBride KM. (2017) . J Exp Med. 214(12):3543-3552.
Ortiz GJ, Li Y, Post SM, Pant V, Xiong S, Larsson CA, El-Naggar AK, Johnson DG, Lozano G. (2017) Oncogene. 37(3):332-340.
Takata KI, Reh S, Yousefzadeh MJ, Zelazowski MJ, Bhetawal S, Trono D, Lowery MG, Sandoval M, Takata Y, Lu Y, Lin K, Shen J, Kusewitt DF, McBride KM, Cole F, Wood RD. (2017) PLoS Genet. 13(6):e1006818.
Schrock MS, Batar B, Lee J, Druck T, Ferguson B, Cho JH, Akakpo K, Hagrass H, Heerema NA, Xia F, Parvin JD, Aldaz CM, Huebner K. (2017) . Oncogene. 2017 Apr 20;36(16):2215-2227.
Veland N, Hardikar S, Zhong Y, Gayatri S, Dan J, Strahl BD, Rothbart SB, Bedford MT, Chen T. (2017) . Cell Rep. 21(12):3390-3397
Wan L*, Wen H*, Li Y, Lyu J, Xi Y, Hoshii T, Joseph JK, Wang X, Loh YE, Erb MA, Souza AL, Bradner JE, Shen L, Li W, Allis CD, Armstrong SA, Shi X. (2017) . Nature. 543(7644):265-269 (*equal contribution)
Zelazowski MJ, Sandoval M, Paniker L, Hamilton HM, Han J, Gribbell MA, Kang R, Cole F. (2017) Cell. 171(3):601-614.e13.
Zhu F, Willette-Brown J, Song NY, Lomada D, Song Y, Xue L, Gray Z, Zhao Z, Davis SR, Sun Z, Zhang P, Wu X, Zhan Q, Richie ER, Hu Y. (2017) . Cell Host Microbe. 21(4):478-493.
2009-2016
2016
Atanassov BS, Mohan RD, Lan X, Kuang X, Lu Y, Lin K, McIvor E, Li W, Zhang Y, Florens L, Byrum SD, Mackintosh SG, Calhoun-Davis T, Koutelou E, Wang L, Tang DG, Tackett AJ, Washburn MP, Workman JL, Dent SY. (2016) . Mol Cell. 62(4):558-71.
Bain VE, Gordon J, O¡¯Neil JD, Ramos I, Richie ER, Manley NR. (2016) . Development. 143(21):4027-4037.
Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, Aronow B, Lin C, Li W, Yang L, Barton MC. (2016) . Mol Cell. 64(5):967-981
Lange SS, Tomida J, Boulware KS, Bhetawal S, Wood RD. (2016) PLoS Genet. 12(1):e1005759.
Li Y, Sabari BR, Panchenko T, Wen H, Zhao D, Guan H, Wan L, Huang H, Tang Z, Zhao Y, Roeder RG, Shi X, Allis CD, Li H. (2016) Mol Cell. 62(2):181-93.
Stuckey JI, Dickson BM, Cheng N, Liu Y, Norris JL, Cholensky SH, Tempel W, Qin S, Huber KG, Sagum C, Black K, Li F, Huang XP, Roth BL, Baughman BM, Senisterra G, Pattenden SG, Vedadi M, Brown PJ, Bedford MT, Min J, Arrowsmith CH, James LI, Frye SV. (2016) . Nat Chem Biol. 12(3):180-7.
V¨¦lez-Cruz R, Manickavinayaham S, Biswas AK, Clary RW, Premkumar T, Cole F, Johnson DG. (2016) . Genes Dev. 2016 Nov 15;30(22):2500-2512.
Wu CC, Lee S, Malladi S, Chen MD, Mastrandrea NJ, Zhang Z, Bratton SB. (2016) . Nat Commun. 2016 Nov 24;7:13565
Zhang D, Park D, Zhong Y, Lu Y, Rycaj K, Gong S, Chen X, Liu X, Chao HP, Whitney P, Calhoun-Davis T, Takata Y, Shen J, Iyer VR, Tang DG. (2016) Nat Commun. 7:10798.
Zhang X, Peng D, Xi Y, Yuan C, Sagum CA, Klein BJ, Tanaka K, Wen H, Kutateladze TG, Li W, Bedford MT, Shi X. (2016) . Nat Commun. 7:10810.
2015
Hirsch CL, Coban Akdemir Z, Wang L, Jayakumaran G, Trcka D, Weiss A, Hernandez JJ, Pan Q, Han H, Xu X, Xia Z, Salinger AP, Wilson M, Vizeacoumar F, Datti A, Li W, Cooney AJ, Barton MC, Blencowe BJ, Wrana JL, Dent SY. (2015) . Genes Dev.29(8):803-16.
Hu SB, Xiang JF, Li X, Xu Y, Xue W, Huang M, Wong CC, Sagum CA, Bedford MT, Yang L, Cheng D, Chen LL. (2015) . Genes Dev. 29(6):630-45.
Kapoor P, Bao Y, Xiao J, Espejo A, Yang L, Bedford MT, Peng G, Shen X. (2015) . Mol Cell. 58(5):863-9.
Kapoor P, Bao Y, Xiao J, Luo J, Shen J, Persinger J, Peng G, Ranish J, Bartholomew B, Shen X. (2015) . Genes Dev. 29(6):591-602.
Li M, Hada A, Sen P, Olufemi L, Hall MA, Smith BY, Forth S, McKnight JN, Patel A, Bowman GD, Bartholomew B, Wang MD. (2015) . Elife. 4. doi: 10.7554/eLife.06249.
Pathiraja TN, Thakkar KN, Jiang S, Stratton S, Liu Z, Gagea M, Shi X, Shah PK, Phan L, Lee MH, Andersen J, Stampfer M, Barton MC. (2015) . Oncogene. 34(22):2836-45.
Yang Y, Hadjikyriacou A, Xia Z, Gayatri S, Kim D, Zurita-Lopez C, Kelly R, Guo A, Li W, Clarke SG, Bedford MT. (2015) . Nat Commun. 6:6428.
2014
Biswas AK, Mitchell DL, Johnson DG (2014) . Cancer Res. 74(12):3369-77.
Cole F, Baudat F, Grey C, Keeney S, de Massy B, Jasin M (2014) . Nat Genet. 46(10):1072-80.
Jin M, Zhang T, Liu C, Badeaux MA, Liu B, Liu R, Jeter C, Chen X, Vlassov AV, Tang DG (2014) . Cancer Res. 74(15):4183-95.
Li Y, Wen H, Xi Y, Tanaka K, Wang H, Peng D, Ren Y, Jin Q, Dent SYR., Li W, Li H, Shi X (2014) Cell.159(3):558-71.
Reeh KA, Cardenas KT, Bain VE, Liu Z, Laurent M, Manley NR, Richie, ER (2014) Development. 141(15):2950-8.
Wen H, Li Y, Xi Y, Jiang S, Stratton S, Peng D, Tanaka K, Ren Y, Xia Z, Wu J, Li B, Barton MC, Li W, Li H, Shi X (2014) Nature. 508(7495):263-8.
Yang Y, McBride KM, Hensley S, Lu Y, Chedin F, Bedford MT (2014) . Mol Cell. 53(3):484-97.
Yousefzadeh MJ, Wyatt DW, Takata K, Mu Y, Hensley SC, Tomida J, Bylund, GO, Doublie S, Johansson E, Ramsden DA, McBride KM and Wood, RD (2014) . PLoS Genet. 10(10):e1004654.
2013
Badeaux MA, Jeter CR, Gong S, Liu B, Suraneni MV, Rundhaug J, Fischer SM, Yang T, Kusewitt D, Tang DG. (2013) . Cell Cycle. 12(15):2395-408.
Chen K, Wilson MA, Hirsch C, Watson A, Liang S, Lu Y, Li W, Dent SY (2013) . Genome Res. 23:312-22.
Johnson DG, Dent SY (2013) . Cell. 152(4):685-9.
Kapoor P, Chen M, Winkler DD, Luger K, Shen X (2013) . Nat Struct Mol Biol. 20(4):426-32.
Kim SJ, Wong PK. (2013) . J Gen Virol. 94(Pt 10):2309-17.
Kim SJ, Zhao H, Hardikar S, Singh AK, Goodell MA, Chen T. (2013) Blood. 2013 Dec 12;122(25):4086-9.
Lange SS, Bedford E, Reh S, Wittschieben JP, Carbajal S, Kusewitt DF, DiGiovanni J, Wood RD (2013) . Proc Natl Acad Sci U S A. 110(8):E687-96.
Li J, Zhao Z, Carter C, Ehrlich LI, Bedford MT, Richie ER (2013) . J Immunol. 190(2):597-604.
Mikulec CD, Rundhaug JE, Simper MS, Lubet RA, Fischer SM. (2013) . Cancer Prev Res (Phila). 6(7):675-85.
Perez CJ, Dumas A, Valli¨¨res L, Gu¨¦net JL, Benavides F. (2013) . J Hered. 2013 Jul-Aug;104(4):565-71.
Takata K, Reh S, Tomida J, Person MD, Wood RD. (2013) . Nat Commun. 2013;4:2338.
Yeh TC, Bratton SB (2013) . Proc Natl Acad Sci U S A. 110(35):14284-9.
Zhao H, Chen T. (2013) . J Hum Genet. 58(7):421-7.
2012
Badeaux AI, Yang Y, Cardenas K, Vemulapalli V, Chen K, Kusewitt D, Richie E, Li W, Bedford MT (2012). . J Biol Chem. 287(1):429-37.
Biswas AK, Johnson DG. (2012) . Cancer Res. 72(1):13-7.
Chen YC, Gatchel JR, Lewis RW, Mao CA, Grant PA, Zoghbi HY, Dent SY (2012) . Hum Mol Genet. 21(2):394-405.
Ferguson BW, Gao X, Kil H, Lee J, Benavides F, Abba MC, Aldaz CM (2012) . PLoS One. 7(5):e36618.
Lange SS, Wittschieben JP, Wood RD (2012) . Nucleic Acids Res. 40 (10):4473-82.
Lu M, Liu Z, Yu H, Wang LE, Li G, Sturgis EM, Johnson DG, Wei Q (2012) . Mol Carcinog. 51 Suppl 1:E132-41.
Mitchell D, Paniker L, Godar D (2012) . Photochem Photobiol. 88(4):1027-32.
Qin J, Liu X, Laffin B, Chen X, Choy G, Jeter CR, Calhoun-Davis T, Li H, Palapattu GS, Pang S, Lin K, Huang J, Ivanov I, Li W, Suraneni MV, Tang DG (2012) Cell Stem Cell. 10(5):556-69.
Regunathan R, Woo J, Pierce MC, Polydorides AD, Raoufi M, Roayaie S, Schwartz M, Labow D, Shin D, Suzuki R, Bhutani MS, Coghlan LG, Richards-Kortum R, Anandasabapathy S, Kim MK. (2012) (with video). Gastrointest Endosc. 76(2):293-300.
2011
Atanassov BS, Dent SY. (2011) EMBO Rep. 12(9):924-30.
Atanassov BS, Koutelou E, Dent SY. (2011) . FEBS Lett. 585(13):2016-23.
Benavides F, Perez C, Blando J, Contreras O, Shen J, Coussens LM, Fischer SM, Kusewitt DF, DiGiovanni J, Conti CJ. (2011) . Mol Carcinog. 51(4):352-61.
Butler JS, Zurita-Lopez CI, Clarke SG, Bedford MT, Dent SY. (2011) . J Biol Chem. 286(14):12234-44.
Chen J, Zhu F, Weaks RL, Biswas AK, Guo R, Li Y, Johnson DG. (2011) . Cell Cycle. 10(8):1287-94.
Fernandez AA, Garcia R, Paniker L, Trono D, Mitchell DL. (2011) . Photochem Photobiol. 87(2):335-41.
Fischer SM, Hawk ET, Lubet RA. (2011) Cancer Prev Res (Phila). 4(11):1728-35.
Guo R, Chen J, Mitchell DL, Johnson DG (2011) . Nucleic Acids Res. 39(4):1390-7.
Jeter CR, Liu B, Liu X, Chen X, Liu C, Calhoun-Davis T, Repass J, Zaehres H, Shen JJ, Tang DG. (2011) . Oncogene. 30(36):3833-45.
Kim J, Hwangbo J, Wong PK. (2011) . PLoS One. 2011 Jan 28;6(1):e16615
Lashinger LM, Malone LM, Brown GW, Daniels EA, Goldberg JA, Otto G, Fischer SM, Hursting SD. (2011) . Cancer Prev Res (Phila). 4(7):1041-51.
Latham JA, Chosed RJ, Wang S, Dent SY (2011) . Cell. 146(5):709-19.
Li H, Tang DG. (2011) . J Surg Oncol. 103(6):558-62.
Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H, Jeter C, Honorio S, Wiggins JF, Bader AG, Fagin R, Brown D, Tang DG (2011) . Nat Med. 17(2):211-5.
Perez C, Parker-Thornburg J, Mikulec C, Kusewitt DF, Fischer SM, Digiovanni J, Conti CJ, Benavides F. (2011) . Exp Dermatol. 21(3):217-20.
Rahn JJ, Rowley B, Lowery MP, Coletta LD, Limanni T, Nairn RS, Adair GM. (2011) . DNA Repair (Amst). 10(2):188-98.
Spannhoff A, Kim YK, Raynal NJ, Gharibyan V, Su MB, Zhou YY, Li J, Castellano S, Sbardella G, Issa JP, Bedford MT (2011) . EMBO Reports. 12(3):238-43.
Surh I, Rundhaug J, Pavone A, Mikulec C, Abel E, Fischer SM. (2011) . Mol Carcinog. 50(6):458-68.
Yan L, Della Coletta L, Powell KL, Shen J, Thames H, Aldaz CM, MacLeod MC. (2011) . PLoS One. 6(1):e16515.
2010
Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, Inoki K, Guan KL, Shen J, Person MD, Kusewitt D, Mills GB, Kastan MB, Walker CL (2010) . Proc Natl Acad Sci U S A.107:4153-8.
Bredfeldt TG, Greathouse KL, Safe SH, Hung MC, Bedford MT, Walker CL. (2010) . Mol Endocrinol. 24(5):993-1006
Jain A, Bacolla A, Chakraborty P, Grosse F, Vasquez KM. (2010) Biochemistry. 49(33):6992-9
Kha DT, Wang G, Natrajan N, Harrison L, Vasquez KM. . J Mol Biol. 2010 398(4):471-80.
Kim D, Lee J, Cheng D, Li J, Carter C, Richie E, Bedford MT. (2010) . J Biol Chem. 285(2):1147-52
Guo R, Chen J, Zhu F, Biswas AK, Berton TR, Mitchell DL, Johnson DG (2010) . J Biol Chem. 285:19308-15.
Mitchell DL, Fernandez AA, Nairn RS, Garcia R, Paniker L, Trono D, Thames HD, Gimenez-Conti I (2010) . Proc Natl Acad Sci U S A. 107(20):9329-34.
Perez CJ, Jaubert J, Gu¨¦net JL, Barnhart KF, Ross-Inta CM, Quintanilla VC, Aubin I, Brandon JL, Otto NW, DiGiovanni J, Gimenez-Conti I, Giulivi C, Kusewitt DF, Conti CJ, Benavides F. (2010) . Am J Pathol. 177(4):1958-68.
Pusapati RV, Weaks RL, Rounbehler RJ, McArthur MJ, Johnson DG. (2010) Mol Carcinog. 49(2):152-6.
Rundhaug JE, Fischer SM. (2010) . Cancers (Basel). 2(2):436-82
Short JD, Dere R, Houston KD, Cai SL, Kim J, Bergeron JM, Shen J, Liang J, Bedford MT, Mills GB, Walker CL. (2010) . Mol Carcinog. 49(5):429-39.
Yang Y, Lu Y, Espejo A, Wu J, Xu W, Liang S, Bedford MT (2010) . Mol Cell. 40(6):1016-23.
Zhu F, Doll¨¦ ME, Berton TR, Kuiper RV, Capps C, Espejo A, McArthur MJ, Bedford MT, van Steeg H, de Vries A, Johnson DG (2010) . Cancer Res. 70(14):5851-9.
2009
Abba MC, Hu Y, Levy CC, Gaddis S, Kittrell FS, Hill J, Bissonnette RP, Brown PH, Medina D, Aldaz CM. (2009) . Cancer Prev Res (Phila). 2(2):175-84.
Blando J, Portis M, Benavides F, Alexander A, Mills G, Dave B, Conti CJ, Kim J, Walker CL. (2009) . Am J Pathol. 174(5):1869-79.
Bua DJ, Kuo AJ, Cheung P, Liu CL, Migliori V, Espejo A, Casadio F, Bassi C, Amati B, Bedford MT, Guccione E, Gozani O. . (2009) PLoS One. 4(8):e6789.
Chen X, Schneider-Broussard R, Hollowell D, McArthur M, Jeter CR, Benavides F, DiGiovanni J, Tang DG. (2009) . Differentiation. 77(3):324-34.
Falbo KB, Alabert C, Katou Y, Wu S, Han J, Wehr T, Xiao J, He X, Zhang Z, Shi Y, Shirahige K, Pasero P, and Shen X (2009) . Nat Struct Mol Biol. 16(11):1167-72.
Griffith AV, Cardenas K, Carter C, Gordon J, Iberg A, Engleka K, Epstein JA, Manley NR, Richie ER (2009) . Dev Biol. 327(1):216-27.
Jeter CR, Badeaux M, Choy G, Chandra D, Patrawala L, Liu C, Calhoun-Davis T, Zaehres H, Daley GQ, Tang DG. (2009) . Stem Cells. 2009 May;27(5):993-1005.
Kim DJ, Kataoka K, Sano S, Connolly K, Kiguchi K, DiGiovanni J. (2009) . Mol Carcinog. 48(10):873-85.
Kuang X, Scofield VL, Yan M, Stoica G, Liu N, Wong PK. (2009) Brain Res. 1286:174-84.
Liu B, Zhu F, Xia X, Park E, Hu Y. (2009) Cell Cycle. 8(4):527-31.
Liu Y, Nairn RS, Vasquez KM. (2009) Nucleic Acids Res. 37(19):6378-88.
Ludes-Meyers JH, Kil H, Parker-Thornburg J, Kusewitt DF, Bedford MT, Aldaz CM. (2009) PLoS One. 4(11):e7775.
Repass JF, Laurent MN, Carter C, Reizis B, Bedford MT, Cardenas K, Narang P, Coles M, Richie ER. (2009) Genesis. 47(4):281-7.
Scofield VL, Yan M, Kuang X, Kim SJ, Crunk D, Wong PK. (2009) Immunol Lett. 122(2):159-69.
Shirley SH, Rundhaug JE, Tian J, Cullinan-Ammann N, Lambertz I, Conti CJ, Fuchs-Young R. (2009) s. Cancer Res. 69(8):3405-14.
Zhao J, Jain A, Iyer RR, Modrich PL, Vasquez KM. (2009) . DNA Repair (Amst). ;8(7):865-72.