Network


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

Hotspot


Dive into the research topics where Mary E. Winn is active.

Publication


Featured researches published by Mary E. Winn.


Breast Cancer: Targets and Therapy | 2015

Critical analysis of the potential for microRNA biomarkers in breast cancer management

Carrie R. Graveel; Heather M. Calderone; Jennifer J Westerhuis; Mary E. Winn; Lorenzo F. Sempere

Breast cancer is a complex and heterogeneous disease. Signaling by estrogen receptor (ER), progesterone receptor (PR), and/or human EGF-like receptor 2 (HER2) is a main driver in the development and progression of a large majority of breast tumors. Molecular characterization of primary tumors has identified major subtypes that correlate with ER/PR/HER2 status, and also subgroup divisions that indicate other molecular and cellular features of the tumors. While some of these research findings have been incorporated into clinical practice, several challenges remain to improve breast cancer management and patient survival, for which the integration of novel biomarkers into current practice should be beneficial. microRNAs (miRNAs) are a class of short non-coding regulatory RNAs with an etiological contribution to breast carcinogenesis. miRNA-based diagnostic and therapeutic applications are rapidly emerging as novel potential approaches to manage and treat breast cancer. Rapid technological development enables specific and sensitive detection of individual miRNAs or the entire miRNome in tissues, blood, and other biological specimens from breast cancer patients. This review focuses on recent miRNA research and its potential to address unmet clinical needs and challenges. The four sections presented discuss miRNA findings in the context of the following clinical challenges: biomarkers for early detection; prognostic and predictive biomarkers for treatment decisions using targeted therapies against ER and HER2; diagnostic and prognostic biomarkers for subgrouping of triple-negative breast cancer, for which there are currently no targeted therapies; and biomarkers for monitoring and characterization of metastatic breast cancer. The review concludes with a critical analysis of the current state of miRNA breast cancer research and the need for further studies using large patient cohorts under well-controlled conditions before considering the clinical implementation of miRNA biomarkers.


American Journal of Pathology | 2014

Stromal Expression of miR-21 Identifies High-Risk Group in Triple-Negative Breast Cancer

Todd A. MacKenzie; Gary N. Schwartz; Heather M. Calderone; Carrie R. Graveel; Mary E. Winn; Galen Hostetter; Wendy A. Wells; Lorenzo F. Sempere

Triple-negative breast cancer (TNBC) is an aggressive subtype defined by the lack of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 expression. Expression of miR-21, an oncomiR, is frequently altered and may be distinctly expressed in the tumor stroma. Because tumor lesions are a complex mixture of cell types, we hypothesized that analysis of miR-21 expression at single-cell resolution could provide more accurate information to assess disease recurrence risk and BC-related death. We implemented a fully automated, tissue slide-based assay to detect miR-21 expression in 988 patients with BC. The miR-21(High) group exhibited shorter recurrence-free survival [hazard ratio (HR), 1.71; P < 0.001] and BC-specific survival (HR, 1.96; P < 0.001) in multivariate regression analyses. When tumor compartment and levels of miR-21 expression were considered, significant associations with poor clinical outcome were detected exclusively in tumor epithelia from estrogen receptor- and/or progesterone receptor-positive human epidermal growth factor receptor 2-negative cases [recurrence-free survival: HR, 3.67 (P = 0.006); BC-specific survival: HR, 5.13 (P = 0.002)] and in tumor stroma from TNBC cases [recurrence-free survival: HR, 2.59 (P = 0.013); BC-specific survival: HR, 3.37 (P = 0.003)]. These findings suggest that the context of altered miR-21 expression provides clinically relevant information. Importantly, miR-21 expression was predominantly up-regulated and potentially prognostic in the tumor stroma of TNBC.


Clinical Cancer Research | 2016

Cabozantinib (XL184) Inhibits Growth and Invasion of Preclinical TNBC Models

Mansoureh Sameni; Elizabeth A. Tovar; Curt Essenburg; Anita Chalasani; Erik S. Linklater; Andrew Borgman; David Cherba; Arulselvi Anbalagan; Mary E. Winn; Carrie R. Graveel; Bonnie F. Sloane

Purpose: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that is associated with poor clinical outcome. There is a vital need for effective targeted therapeutics for TNBC patients, yet treatment strategies are challenged by the significant intertumoral heterogeneity within the TNBC subtype and its surrounding microenvironment. Receptor tyrosine kinases (RTK) are highly expressed in several TNBC subtypes and are promising therapeutic targets. In this study, we targeted the MET receptor, which is highly expressed across several TNBC subtypes. Experimental Design: Using the small-molecule inhibitor cabozantinib (XL184), we examined the efficacy of MET inhibition in preclinical models that recapitulate human TNBC and its microenvironment. To analyze the dynamic interactions between TNBC cells and fibroblasts over time, we utilized a 3D model referred to as MAME (Mammary Architecture and Microenvironment Engineering) with quantitative image analysis. To investigate cabozantinib inhibition in vivo, we used a novel xenograft model that expresses human HGF and supports paracrine MET signaling. Results: XL184 treatment of MAME cultures of MDA-MB-231 and HCC70 cells (± HGF-expressing fibroblasts) was cytotoxic and significantly reduced multicellular invasive outgrowths, even in cultures with HGF-expressing fibroblasts. Treatment with XL184 had no significant effects on METneg breast cancer cell growth. In vivo assays demonstrated that cabozantinib treatment significantly inhibited TNBC growth and metastasis. Conclusions: Using preclinical TNBC models that recapitulate the breast tumor microenvironment, we demonstrate that cabozantinib inhibition is an effective therapeutic strategy in several TNBC subtypes. Clin Cancer Res; 22(4); 923–34. ©2015 AACR.


Clinical Cancer Research | 2016

Identification of Mithramycin Analogues with Improved Targeting of the EWS-FLI1 Transcription Factor

Christy L. Osgood; Nichole Maloney; Christopher G. Kidd; Susan M. Kitchen-Goosen; Laura E. Segars; Meti Gebregiorgis; Girma M. Woldemichael; Min He; Savita Sankar; Stephen L. Lessnick; Min H. Kang; Malcolm A. Smith; Lisa Turner; Zachary Madaj; Mary E. Winn; Luz-Elena Núñez; Javier González-Sabín; Lee J. Helman; Francisco Morís; Patrick J. Grohar

Purpose: The goal of this study was to identify second-generation mithramycin analogues that better target the EWS-FLI1 transcription factor for Ewing sarcoma. We previously established mithramycin as an EWS-FLI1 inhibitor, but the compounds toxicity prevented its use at effective concentrations in patients. Experimental Design: We screened a panel of mithralogs to establish their ability to inhibit EWS-FLI1 in Ewing sarcoma. We compared the IC50 with the MTD established in mice to determine the relationship between efficacy and toxicity. We confirmed the suppression of EWS-FLI1 at the promoter, mRNA, gene signature, and protein levels. We established an improved therapeutic window by using time-lapse microscopy to model the effects on cellular proliferation in Ewing sarcoma cells relative to HepG2 control cells. Finally, we established an improved therapeutic window using a xenograft model of Ewing sarcoma. Results: EC-8105 was found to be the most potent analogue and was able to suppress EWS-FLI1 activity at concentrations nontoxic to other cell types. EC-8042 was substantially less toxic than mithramycin in multiple species but maintained suppression of EWS-FLI1 at similar concentrations. Both compounds markedly suppressed Ewing sarcoma xenograft growth and inhibited EWS-FLI1 in vivo. Conclusions: These results provide a basis for the continued development of EC-8042 and EC-8105 as EWS-FLI1 inhibitors for the clinic. Clin Cancer Res; 22(16); 4105–18. ©2016 AACR.


Nature Communications | 2017

The genomic landscape of tuberous sclerosis complex

Katie R. Martin; Wanding Zhou; Megan J. Bowman; Juliann Shih; Kit Sing Au; Kristin E. Dittenhafer-Reed; Kellie A. Sisson; Julie Koeman; Daniel J. Weisenberger; Sandra Cottingham; Steven T. DeRoos; Orrin Devinsky; Mary E. Winn; Andrew D. Cherniack; Hui Shen; Hope Northrup; Darcy A. Krueger; Jeffrey P. MacKeigan

Tuberous sclerosis complex (TSC) is a rare genetic disease causing multisystem growth of benign tumours and other hamartomatous lesions, which leads to diverse and debilitating clinical symptoms. Patients are born with TSC1 or TSC2 mutations, and somatic inactivation of wild-type alleles drives MTOR activation; however, second hits to TSC1/TSC2 are not always observed. Here, we present the genomic landscape of TSC hamartomas. We determine that TSC lesions contain a low somatic mutational burden relative to carcinomas, a subset feature large-scale chromosomal aberrations, and highly conserved molecular signatures for each type exist. Analysis of the molecular signatures coupled with computational approaches reveals unique aspects of cellular heterogeneity and cell origin. Using immune data sets, we identify significant neuroinflammation in TSC-associated brain tumours. Taken together, this molecular catalogue of TSC serves as a resource into the origin of these hamartomas and provides a framework that unifies genomic and transcriptomic dimensions for complex tumours.


American Journal of Medical Genetics Part A | 2015

Oculoectodermal syndrome is a mosaic RASopathy associated with KRAS alterations.

Jacqueline D. Peacock; Karl Dykema; Helga V. Toriello; Marie R. Mooney; Donald J. Scholten; Mary E. Winn; Andrew Borgman; Nicholas S. Duesbery; Judith A. Hiemenga; Cong Liu; Stacey Campbell; Brian P. Nickoloff; Bart O. Williams; Matthew R. Steensma

Oculoectodermal syndrome (OES) is a rare disease characterized by a combination of congenital scalp lesions and ocular dermoids, with additional manifestations including non‐ossifying fibromas and giant cell granulomas of the jaw occurring during the first decade of life. To identify the genetic etiology of OES, we conducted whole‐genome sequencing of several tissues in an affected individual. Comparison of DNA from a non‐ossifying fibroma to blood‐derived DNA allowed identification of a somatic missense alteration in KRAS NM_033360.3(KRAS):c.38G>A, resulting in p.Gly13Asp. This alteration was also observed in the patients other affected tissues including the skin and muscle. Targeted sequencing in a second, unrelated OES patient identified an NM_033360.3(KRAS):c.57G>C, p.Leu19Phe alteration. Allelic frequencies fell below 40% in all tissues examined in both patients, suggesting that OES is a mosaic RAS‐related disorder, or RASopathy. The characteristic findings in OES, including scalp lesions, ocular dermoids, and benign tumors, are found in other mosaic and germline RASopathies. This discovery also broadens our understanding of the spectrum of phenotypes resulting from KRAS alterations. Future research into disease progression with regard to malignancy risk and investigation of RAS‐targeted therapies in OES is warranted. KRAS sequencing is clinically available and may also now improve OES diagnostic criteria.


Oncotarget | 2016

Targeting MET and EGFR crosstalk signaling in triple-negative breast cancers.

Erik S. Linklater; Elizabeth A. Tovar; Curt Essenburg; Lisa Turner; Zachary Madaj; Mary E. Winn; Marianne Melnik; Hasan Korkaya; Christiane R. Maroun; James G. Christensen; Matthew R. Steensma; Julie L. Boerner; Carrie R. Graveel

There is a vital need for improved therapeutic strategies that are effective in both primary and metastatic triple-negative breast cancer (TNBC). Current treatment options for TNBC patients are restricted to chemotherapy; however tyrosine kinases are promising druggable targets due to their high expression in multiple TNBC subtypes. Since coexpression of receptor tyrosine kinases (RTKs) can promote signaling crosstalk and cell survival in the presence of kinase inhibitors, it is likely that multiple RTKs will need to be inhibited to enhance therapeutic benefit and prevent resistance. The MET and EGFR receptors are actionable targets due to their high expression in TNBC; however crosstalk between MET and EGFR has been implicated in therapeutic resistance to single agent use of MET or EGFR inhibitors in several cancer types. Therefore it is likely that dual inhibition of MET and EGFR is required to prevent crosstalk signaling and acquired resistance. In this study, we evaluated the heterogeneity of MET and EGFR expression and activation in primary and metastatic TNBC tumorgrafts and determined the efficacy of MET (MGCD265 or crizotinib) and/or EGFR (erlotinib) inhibition against TNBC progression. Here we demonstrate that combined MET and EGFR inhibition with either MGCD265 and erlotinib treatment or crizotinib and erlotinib treatment were highly effective at abrogating tumor growth and significantly decreased the variability in treatment response compared to monotherapy. These results advance our understanding of the RTK signaling architecture in TNBC and demonstrate that combined MET and EGFR inhibition may be a promising therapeutic strategy for TNBC patients.


Free Radical Biology and Medicine | 2017

Uncoupling of oxidative stress resistance and lifespan in long-lived isp-1 mitochondrial mutants in Caenorhabditis elegans

Dylan J. Dues; Claire E. Schaar; Benjamin K. Johnson; Megan J. Bowman; Mary E. Winn; Megan M. Senchuk; Jeremy M. Van Raamsdonk

Abstract Mutations affecting components of the mitochondrial electron transport chain have been shown to increase lifespan in multiple species including the worm Caenorhabditis elegans. While it was originally proposed that decreased generation of reactive oxygen species (ROS) resulting from lower rates of electron transport could account for the observed increase in lifespan, recent evidence indicates that ROS levels are increased in at least some of these long‐lived mitochondrial mutants. Here, we show that the long‐lived mitochondrial mutant isp‐1 worms have increased resistance to oxidative stress. Our results suggest that elevated ROS levels in isp‐1 worms cause the activation of multiple stress‐response pathways including the mitochondrial unfolded protein response, the SKN‐1‐mediated stress response, and the hypoxia response. In addition, these worms have increased expression of specific antioxidant enzymes, including a marked upregulation of the inducible superoxide dismutase genes sod‐3 and sod‐5. Examining the contribution of sod‐3 and sod‐5 to the oxidative stress resistance in isp‐1 worms revealed that loss of either of these genes increased resistance to oxidative stress, but not other forms of stress. Deletion of sod‐3 or sod‐5 decreased the lifespan of isp‐1 worms and further exacerbated their slow physiologic rates. Thus, while deletion of sod‐3 and sod‐5 genes has little impact on stress resistance, physiologic rates or lifespan in wild‐type worms, these genes are required for the longevity of isp‐1 worms. Overall, this work shows that the increased resistance to oxidative stress in isp‐1 worms does not account for their longevity, and that resistance to oxidative stress can be experimentally dissociated from lifespan. Graphical abstract Figure. No Caption available. Highlightsisp‐1 worms have increased resistance to oxidative stress despite having elevated levels of ROS.Deletion of inducible superoxide dismutase genes paradoxically increases resistance to oxidative stress.Inducible superoxide dismutase genes can impact longevity, stress resistance and physiologic rates.Resistance to oxidative stress can be experimentally dissociated from longevity.


Nature Genetics | 2018

Discordant inheritance of chromosomal and extrachromosomal DNA elements contributes to dynamic disease evolution in glioblastoma.

Ana C. deCarvalho; Hoon Kim; Laila M. Poisson; Mary E. Winn; Claudius Mueller; David Cherba; Julie Koeman; Sahil Seth; Alexei Protopopov; Michelle Felicella; Siyuan Zheng; Asha S. Multani; Yongying Jiang; Jianhua Zhang; Do Hyun Nam; Emanuel F. Petricoin; Lynda Chin; Tom Mikkelsen; Roel G.W. Verhaak

To understand how genomic heterogeneity of glioblastoma (GBM) contributes to poor therapy response, we performed DNA and RNA sequencing on GBM samples and the neurospheres and orthotopic xenograft models derived from them. We used the resulting dataset to show that somatic driver alterations including single-nucleotide variants, focal DNA alterations and oncogene amplification on extrachromosomal DNA (ecDNA) elements were in majority propagated from tumor to model systems. In several instances, ecDNAs and chromosomal alterations demonstrated divergent inheritance patterns and clonal selection dynamics during cell culture and xenografting. We infer that ecDNA was unevenly inherited by offspring cells, a characteristic that affects the oncogenic potential of cells with more or fewer ecDNAs. Longitudinal patient tumor profiling found that oncogenic ecDNAs are frequently retained throughout the course of disease. Our analysis shows that extrachromosomal elements allow rapid increase of genomic heterogeneity during GBM evolution, independently of chromosomal DNA alterations.Analysis of glioblastoma samples and derived neurospheres and xenografts shows that chromosomal and extrachromosomal alterations often display divergent inheritance patterns during cell culture and xenografting.


Frontiers in Plant Science | 2016

A Developmental and Molecular View of Formation of Auxin-Induced Nodule-Like Structures in Land Plants

Ryan Hiltenbrand; Jacklyn Thomas; Hannah McCarthy; Karl Dykema; Ashley Spurr; Hamilton Newhart; Mary E. Winn; Arijit Mukherjee

Several studies have shown that plant hormones play important roles during legume–rhizobia symbiosis. For instance, auxins induce the formation of nodule-like structures (NLSs) on legume roots in the absence of rhizobia. Furthermore, these NLS can be colonized by nitrogen-fixing bacteria, which favor nitrogen fixation compared to regular roots and subsequently increase plant yield. Interestingly, auxin also induces similar NLS in cereal roots. While several genetic studies have identified plant genes controlling NLS formation in legumes, no studies have investigated the genes involved in NLS formation in cereals. In this study, first we established an efficient experimental system to induce NLS in rice roots, using auxin, 2,4-D, consistently at a high frequency (>90%). We were able to induce NLS at a high frequency in Medicago truncatula under similar conditions. NLS were characterized by a broad base, a diffuse meristem, and increased cell differentiation in the vasculature. Interestingly, NLS formation appeared very similar in both rice and Medicago, suggesting a similar developmental program. We show that NLS formation in both rice and Medicago occurs downstream of the common symbiotic pathway. Furthermore, NLS formation occurs downstream of cytokinin-induced step(s). We performed a comprehensive RNA sequencing experiment to identify genes differentially expressed during NLS formation in rice and identified several promising genes for control of NLS based on their biological and molecular functions. We validated the expression patterns of several genes using reverse transcription polymerase chain reaction and show varied expression patterns of these genes during different stages of NLS formation. Finally, we show that NLS induced on rice roots under these conditions can be colonized by nitrogen-fixing bacteria, Azorhizobium caulinodans.

Collaboration


Dive into the Mary E. Winn's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge