Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jeffrey S. Damrauer is active.

Publication


Featured researches published by Jeffrey S. Damrauer.


Journal of Clinical Investigation | 2004

Cancer cachexia is regulated by selective targeting of skeletal muscle gene products

Swarnali Acharyya; Katherine J. Ladner; Lori L. Nelsen; Jeffrey S. Damrauer; Peter J. Reiser; Steven Swoap; Denis C. Guttridge

Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology

Jeffrey S. Damrauer; Katherine A. Hoadley; David D. Chism; Cheng Fan; Christopher J. Tiganelli; Sara E. Wobker; Jen Jen Yeh; Matthew I. Milowsky; Gopa Iyer; Joel S. Parker; William Y. Kim

Significance The identification of molecular subtype heterogeneity in breast cancer has allowed a deeper understanding of breast cancer biology. We present evidence that there are two intrinsic subtypes of high-grade bladder cancer, basal-like and luminal, which reflect the hallmarks of breast biology. Moreover, we have developed an accurate gene set predictor of molecular subtype, the BASE47, that should allow the incorporation of subtype stratification into clinical trials. Further clinical, etiologic, and therapeutic response associations will be of interest in future investigations. We sought to define whether there are intrinsic molecular subtypes of high-grade bladder cancer. Consensus clustering performed on gene expression data from a meta-dataset of high-grade, muscle-invasive bladder tumors identified two intrinsic, molecular subsets of high-grade bladder cancer, termed “luminal” and “basal-like,” which have characteristics of different stages of urothelial differentiation, reflect the luminal and basal-like molecular subtypes of breast cancer, and have clinically meaningful differences in outcome. A gene set predictor, bladder cancer analysis of subtypes by gene expression (BASE47) was defined by prediction analysis of microarrays (PAM) and accurately classifies the subtypes. Our data demonstrate that there are at least two molecularly and clinically distinct subtypes of high-grade bladder cancer and validate the BASE47 as a subtype predictor. Future studies exploring the predictive value of the BASE47 subtypes for standard of care bladder cancer therapies, as well as novel subtype-specific therapies, will be of interest.


Nature Communications | 2015

Coexistent ARID1A–PIK3CA mutations promote ovarian clear-cell tumorigenesis through pro-tumorigenic inflammatory cytokine signalling

Ronald L. Chandler; Jeffrey S. Damrauer; Jesse R. Raab; Jonathan C. Schisler; Matthew D. Wilkerson; John P. Didion; Joshua Starmer; Daniel W. Serber; Della Yee; Jessie Xiong; David B. Darr; Fernando Pardo-Manuel de Villena; William Y. Kim; Terry Magnuson

Ovarian clear-cell carcinoma (OCCC) is an aggressive form of ovarian cancer with high ARID1A mutation rates. Here we present a mutant mouse model of OCCC. We find that ARID1A inactivation is not sufficient for tumor formation, but requires concurrent activation of the phosphoinositide 3-kinase catalytic subunit, PIK3CA. Remarkably, the mice develop highly penetrant tumors with OCCC-like histopathology, culminating in hemorrhagic ascites and a median survival period of 7.5 weeks. Therapeutic treatment with the pan-PI3K inhibitor, BKM120, prolongs mouse survival by inhibiting tumor cell growth. Cross-species gene expression comparisons support a role for IL-6 inflammatory cytokine signaling in OCCC pathogenesis. We further show that ARID1A and PIK3CA mutations cooperate to promote tumor growth through sustained IL-6 overproduction. Our findings establish an epistatic relationship between SWI/SNF chromatin remodeling and PI3K pathway mutations in OCCC and demonstrate that these pathways converge on pro-tumorigenic cytokine signaling. We propose that ARID1A protects against inflammation-driven tumorigenesis.


Journal of Clinical Investigation | 2014

Erythropoietin promotes breast tumorigenesis through tumor-initiating cell self-renewal.

Bing Zhou; Jeffrey S. Damrauer; Sean T. Bailey; Tanja Hadzic; Youngtae Jeong; Kelly S. Clark; Cheng Fan; Laura Murphy; Cleo Lee; Melissa A. Troester; C. Ryan Miller; Jian Jin; David B. Darr; Charles M. Perou; Ross L. Levine; Maximilian Diehn; William Y. Kim

Erythropoietin (EPO) is a hormone that induces red blood cell production. In its recombinant form, EPO is the one of most prescribed drugs to treat anemia, including that arising in cancer patients. In randomized trials, EPO administration to cancer patients has been associated with decreased survival. Here, we investigated the impact of EPO modulation on tumorigenesis. Using genetically engineered mouse models of breast cancer, we found that EPO promoted tumorigenesis by activating JAK/STAT signaling in breast tumor-initiating cells (TICs) and promoted TIC self renewal. We determined that EPO was induced by hypoxia in breast cancer cell lines, but not in human mammary epithelial cells. Additionally, we demonstrated that high levels of endogenous EPO gene expression correlated with shortened relapse-free survival and that pharmacologic JAK2 inhibition was synergistic with chemotherapy for tumor growth inhibition in vivo. These data define an active role for endogenous EPO in breast cancer progression and breast TIC self-renewal and reveal a potential application of EPO pathway inhibition in breast cancer therapy.


PLOS ONE | 2014

mTOR inhibition induces compensatory, therapeutically targetable MEK activation in renal cell carcinoma.

Sean T. Bailey; Bing Zhou; Jeffrey S. Damrauer; Bhavani Krishnan; Harper L. Wilson; Aleisha M. Smith; Mingqing Li; Jen Jen Yeh; William Y. Kim

Rapamycin derivatives allosterically targeting mTOR are currently FDA approved to treat advanced renal cell carcinoma (RCC), and catalytic inhibitors of mTOR/PI3K are now in clinical trials for treating various solid tumors. We sought to investigate the relative efficacy of allosteric versus catalytic mTOR inhibition, evaluate the crosstalk between the mTOR and MEK/ERK pathways, as well as the therapeutic potential of dual mTOR and MEK inhibition in RCC. Pharmacologic (rapamycin and BEZ235) and genetic manipulation of the mTOR pathway were evaluated by in vitro assays as monotherapy as well as in combination with MEK inhibition (GSK1120212). Catalytic mTOR inhibition with BEZ235 decreased proliferation and increased apoptosis better than allosteric mTOR inhibition with rapamycin. While mTOR inhibition upregulated MEK/ERK signaling, concurrent inhibition of both pathways had enhanced therapeutic efficacy. Finally, primary RCC tumors could be classified into subgroups [(I) MEK activated, (II) Dual MEK and mTOR activated, (III) Not activated, and (IV) mTOR activated] based on their relative activation of the PI3K/mTOR and MEK pathways. Patients with mTOR only activated tumors had the worst prognosis. In summary, dual targeting of the mTOR and MEK pathways in RCC can enhance therapeutic efficacy and primary RCC can be subclassified based on their relative levels of mTOR and MEK activation with potential therapeutic implications.


European Journal of Translational Myology | 2018

Chemotherapy-induced muscle wasting: association with NF-κB and cancer cachexia

Jeffrey S. Damrauer; Michael E. Stadler; Swarnali Acharyya; Albert S. Baldwin; Marion E. Couch; Denis C. Guttridge

A compounding feature of greater than 50% of all cancers is the high incidence of the cachexia syndrome, a complex metabolic disorder characterized by extreme weight loss due mainly to the gross depletion of skeletal muscle tissue. Although studies into the cause of cancer cachexia has spanned over multiple decades, little is known about the effects of various cancer treatments themselves on cachexia. For example, chemotherapy agents induce side effects such as nausea and anorexia, but these symptoms do not fully account for the changes seen with cancer cachexia. In this study we examine the effects of chemotherapeutic compounds, specifically, cisplatin in the colon-26 adenocarcinoma model of cancer cachexia. We find that although cisplatin is able to reduce tumor burden as expected, muscle wasting in mice nevertheless persists. Strikingly, cisplatin alone was seen to regulate muscle atrophy, which was independent of the commonly implicated ubiquitin proteasome system. Finally, we show that cisplatin is able to induce NF-κB activity in both mouse muscles and myotube cultures, suggesting that an additional side effect of cancer treatment is the regulation of muscle wasting that may be mediated through activation of the NF-κB signaling pathway.


Cancer Research | 2018

Molecular subtype-specific immunocompetent models of high-grade urothelial carcinoma reveal differential neoantigen expression and response to immunotherapy

Ryoichi Saito; Christof C. Smith; Takanobu Utsumi; Lisa M. Bixby; Jordan Kardos; Sara E. Wobker; Kyle Stewart; Shengjie Chai; Ujjawal Manocha; Kevin M. Byrd; Jeffrey S. Damrauer; Scott E. Williams; Benjamin G. Vincent; William Y. Kim

High-grade urothelial cancer contains intrinsic molecular subtypes that exhibit differences in underlying tumor biology and can be divided into luminal-like and basal-like subtypes. We describe here the first subtype-specific murine models of bladder cancer and show that Upk3a-CreERT2; Trp53L/L; PtenL/L; Rosa26LSL-Luc (UPPL, luminal-like) and BBN (basal-like) tumors are more faithful to human bladder cancer than the widely used MB49 cells. Following engraftment into immunocompetent C57BL/6 mice, BBN tumors were more responsive to PD-1 inhibition than UPPL tumors. Responding tumors within the BBN model showed differences in immune microenvironment composition, including increased ratios of CD8+:CD4+ and memory:regulatory T cells. Finally, we predicted and confirmed immunogenicity of tumor neoantigens in each model. These UPPL and BBN models will be a valuable resource for future studies examining bladder cancer biology and immunotherapy.Significance: This work establishes human-relevant mouse models of bladder cancer. Cancer Res; 78(14); 3954-68. ©2018 AACR.


Cancer Research | 2017

Abstract 1654: Development of subtype specific mouse models of bladder cancer

Christof C. Smith; Ryoichi Saito; Lisa M. Bixby; Takanobu Utsumi; Jordan Kardos; Shengjie Chai; Sara E. Wobker; Bhavani Krishnan; Jeffrey S. Damrauer; Jonathan S. Serody; David B. Darr; Benjamin G. Vincent; William Y. Kim

Introduction: High-grade, muscle-invasive bladder cancer has recently been shown to harbor intrinsic molecular subtypes with distinct biologic features. Current murine models of bladder cancer, including the prominent carcinogen induced model MB49, do not account for subtype specific characteristics, leaving a gap in available tools for understanding subtype specific differences in bladder cancer. We have developed and validated immunocompetent, subtype specific models of bladder cancer, and we have used these models to assess differential responses to immune checkpoint inhibition. Methods: Two distinct models of murine bladder cancer were developed in a C57BL/6 background. The UPPL models were generated through Pten/Trp53 conditional knockout in Uroplakin3a expressing cells. BBN models were generated through exposure of wild-type C57BL/6 mice to the carcinogen N-Butyl-N-(4-hydmoxybutyl)nitrosamine and subsequent generation of cell lines from spontaneous tumors. RNAseq was performed on several BBN and UPPL tumors and cell lines, with findings validated with flow cytometry and T/B cell receptor (TCR/BCR) amplicon sequencing of tumor infiltrating lymphocytes (TILs). Results: BBN and UPPL models reflected characteristics of human basal and luminal bladder cancers, respectively. BBN (basal) models demonstrated higher immune gene signature expression, with concordantly higher numbers of TILs compared to the UPPL (luminal) model (p Discussion: We have developed two unique classes of murine bladder cancer lines, UPPL and BBN, with gene expression and TIL profiles that closely correlate with human luminal and basal bladder cancers, respectively. The BBN and UPPL subtype specific models can serve as a tool for elucidating bladder cancer responses to immunotherapy. The mixed response of BBN963 tumors to PD-1 blockade should be an asset for assessing pathways mediating response to checkpoint blockade as well as the value of combination therapy. [C.S., R.S, B.V, W.K contributed equally to this work] Citation Format: Christof C. Smith, Ryoichi Saito, Lisa M. Bixby, Takanobu Utsumi, Jordan Kardos, Shengjie Chai, Sara E. Wobker, Bhavani Krishnan, Jeffrey S. Damrauer, Jonathan S. Serody, David Darr, Benjamin G. Vincent, William Y. Kim. Development of subtype specific mouse models of bladder cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1654. doi:10.1158/1538-7445.AM2017-1654


Bioinformatics | 2018

bcSeq: an R package for fast sequence mapping in high-throughput shRNA and CRISPR screens

Jiaxing Lin; Jeremy Gresham; Tongrong Wang; So Young Kim; James V. Alvarez; Jeffrey S. Damrauer; Scott R. Floyd; Joshua A. Granek; Andrew S. Allen; Cliburn Chan; Jichun Xie; Kouros Owzar

Summary: CRISPR‐Cas9 and shRNA high‐throughput sequencing screens have abundant applications for basic and translational research. Methods and tools for the analysis of these screens must properly account for sequencing error, resolve ambiguous mappings among similar sequences in the barcode library in a statistically principled manner, and be computationally efficient. Herein we present bcSeq, an open source R package that implements a fast and parallelized algorithm for mapping high‐throughput sequencing reads to a barcode library while tolerating sequencing error. The algorithm uses a Trie data structure for speed and resolves ambiguous mappings by using a statistical sequencing error model based on Phred scores for each read. Availability and implementation: The package source code and an accompanying tutorial are available at http://bioconductor.org/packages/bcSeq/. Supplementary information: Supplementary data are available at Bioinformatics online.


The Journal of Urology | 2017

MP88-20 ESTABLISHMENT OF NOVEL MOUSE BLADDER CANCER CELL LINES MIMICKING INTRINSIC SUBTYPE OF HUMAN INVASIVE BLADDER CANCER

Ryoichi Saito; Christof C. Smith; Jordan Kardos; Lisa M. Bixby; Shengjie Chai; Jeffrey S. Damrauer; Takanobu Utsumi; Sara E. Wobker; Bhavani Krishnan; Osamu Ogawa; Benjamin G. Vincent; William Y. Kim

experiments such as Western blotting analysis(WB), Hoechst33342 staining and immunostaining. The evaluation of targeting KLF4 by miR145 was performed by luciferase assay. Moreover, the effects of knockdown of KLF4 on the various phenotypes of BC cells were also examined. Furthermore, the networks involving KLF4/PTBP1/PKMs in the Warburg effect relatedmolecules were examined by WB even in clinical BC samples. Finally, we examined immunohistochemical staining to evaluate KLF4 expression. RESULTS: The expression levels of miR-145 were significantly down-regulated in clinical BC samples and BC cells compared with those in normal tissues and HUC. Luciferase assay showed that miR-145 directly targeted KLF4. Also, the Warburg related affectgenes were modulated by the transfection of miR-145 or siR-KLF4 in BC cells. Moreover, the expression levels of KLF4, PTBP1, and PKM2 is up-regulated in all clinical samples by WB. Finally, we observed KLF4-positive staining in the tumor by immunohistochemical staining. CONCLUSIONS: In this study, we indicated that miR-145 affect the Warburg effect through targeting KLF4/PTBP1/PKMs axis in BC cells, which exhibited a significant cell growth inhibition.

Collaboration


Dive into the Jeffrey S. Damrauer's collaboration.

Top Co-Authors

Avatar

William Y. Kim

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Katherine A. Hoadley

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Sara E. Wobker

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin G. Vincent

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chad J. Creighton

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Christof C. Smith

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

David B. Darr

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Dean F. Bajorin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge