Network


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

Hotspot


Dive into the research topics where Patrick M. Woster is active.

Publication


Featured researches published by Patrick M. Woster.


Nature Medicine | 2012

Inhibition of the LSD1 (KDM1A) demethylase reactivates the all- trans -retinoic acid differentiation pathway in acute myeloid leukemia

Tino Schenk; Weihsu Claire Chen; Stefanie Göllner; Louise Howell; Liqing Jin; Katja Hebestreit; Hans-Ulrich Klein; Andreea C. Popescu; Alan Kenneth Burnett; Ken I. Mills; Robert A. Casero; Laurence J. Marton; Patrick M. Woster; Mark D. Minden; Martin Dugas; Jean C.Y. Wang; John E. Dick; Carsten Müller-Tidow; Kevin Petrie; Arthur Zelent

Acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML), characterized by the t(15;17)-associated PML-RARA fusion, has been successfully treated with therapy utilizing all-trans-retinoic acid (ATRA) to differentiate leukemic blasts. However, among patients with non-APL AML, ATRA-based treatment has not been effective. Here we show that, through epigenetic reprogramming, inhibitors of lysine-specific demethylase 1 (LSD1, also called KDM1A), including tranylcypromine (TCP), unlocked the ATRA-driven therapeutic response in non-APL AML. LSD1 inhibition did not lead to a large-scale increase in histone 3 Lys4 dimethylation (H3K4me2) across the genome, but it did increase H3K4me2 and expression of myeloid-differentiation–associated genes. Notably, treatment with ATRA plus TCP markedly diminished the engraftment of primary human AML cells in vivo in nonobese diabetic (NOD)-severe combined immunodeficient (SCID) mice, suggesting that ATRA in combination with TCP may target leukemia-initiating cells. Furthermore, initiation of ATRA plus TCP treatment 15 d after engraftment of human AML cells in NOD-SCID γ (with interleukin-2 (IL-2) receptor γ chain deficiency) mice also revealed the ATRA plus TCP drug combination to have a potent anti-leukemic effect that was superior to treatment with either drug alone. These data identify LSD1 as a therapeutic target and strongly suggest that it may contribute to AML pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Inhibition of lysine-specific demethylase 1 by polyamine analogues results in reexpression of aberrantly silenced genes

Yi Huang; Eriko Greene; Tracy Murray Stewart; Andrew C. Goodwin; Stephen B. Baylin; Patrick M. Woster; Robert A. Casero

Epigenetic chromatin modification is a major regulator of eukaryotic gene expression, and aberrant epigenetic silencing of gene expression contributes to tumorigenesis. Histone modifications include acetylation, phosphorylation, and methylation, resulting in a combination of histone marks known collectively as the histone code. The chromatin marks at a given promoter determine, in part, whether specific promoters are in an open/active conformation or closed/repressed conformation. Dimethyl-lysine 4 histone H3 (H3K4me2) is a transcription-activating chromatin mark at gene promoters, and demethylation of this mark by the lysine-specific demethylase 1 (LSD1), a homologue of polyamine oxidases, may broadly repress gene expression. We now report that novel biguanide and bisguanidine polyamine analogues are potent inhibitors of LSD1. These analogues inhibit LSD1 in human colon carcinoma cells and affect a reexpression of multiple, aberrantly silenced genes important in the development of colon cancer, including members of the secreted frizzle-related proteins (SFRPs) and the GATA family of transcription factors. Furthermore, we demonstrate by chromatin immunoprecipitation analysis that the reexpression is concurrent with increased H3K4me2 and acetyl-H3K9 marks, decreased H3K9me1 and H3K9me2 repressive marks. We thus define important new agents for reversing aberrant repression of gene transcription.


Clinical Pharmacology & Therapeutics | 1999

6′,7′‐Dihydroxybergamottin in grapefruit juice and Seville orange juice: Effects on cyclosporine disposition, enterocyte CYP3A4, and P‐glycoprotein

David J. Edwards; Michael E. Fitzsimmons; Erin G. Schuetz; Kazuto Yasuda; Murray P. Ducharme; Lawrence Warbasse; Patrick M. Woster; John D. Schuetz; Paul B. Watkins

6′,7′‐Dihydroxybergamottin is a furanocoumarin that inhibits CYP3A4 and is found in grapefruit juice and Seville orange juice. Grapefruit juice increases the oral bioavailability of many CYP3A4 substrates, including cyclosporine (INN, ciclosporin), but intestinal P‐glycoprotein may be a more important determinant of cyclosporine availability.


Clinical Cancer Research | 2009

Novel Oligoamine Analogues Inhibit Lysine-Specific Demethylase 1 and Induce Reexpression of Epigenetically Silenced Genes

Yi Huang; Tracy Murray Stewart; Yu Wu; Stephen B. Baylin; Laurence J. Marton; Brandy Perkins; Richard J. Jones; Patrick M. Woster; Robert A. Casero

Purpose: Abnormal DNA CpG island hypermethylation and transcriptionally repressive histone modifications are associated with the aberrant silencing of tumor suppressor genes. Lysine methylation is a dynamic, enzymatically controlled process. Lysine-specific demethylase 1 (LSD1) has recently been identified as a histone lysine demethylase. LSD1 specifically catalyzes demethylation of mono– and dimethyl–lysine 4 of histone 3 (H3K4), key positive chromatin marks associated with transcriptional activation. We hypothesized that a novel class of oligoamine analogues would effectively inhibit LSD1 and thus cause the reexpression of aberrantly silenced genes. Experimental Design: Human colorectal cancer cells were treated with the oligoamines and changes in mono- and dimethyl-H3K4 and other chromatin marks were monitored. In addition, treated cells were evaluated for the reexpression of the aberrantly silenced secreted frizzled-related proteins (SFRP) Wnt signaling pathway antagonist genes. Finally, the effects of the LSD1 inhibitors were evaluated in an in vivo xenograft model. Results: Treatment of HCT116 human colon adenocarcinoma cells in vitro resulted in increased H3K4 methylation and reexpression of silenced SFRP genes. This reexpression is also accompanied by a decrease in H3K9me2 repressive mark. Importantly, cotreatment with low doses of oligoamines and a DNA methyltransferase inhibitor highly induces the reexpression of the aberrantly silenced SFRP2 gene and results in significant inhibition of the growth of established tumors in a human colon tumor model in vivo. Conclusions: The use of LSD1-inhibiting oligoamine analogues in combination with DNA methyltransferase inhibitors represents a highly promising and novel approach for epigenetic therapy of cancer. (Clin Cancer Res 2009;15(23):7217–28)


Amino Acids | 2007

Spermine and spermidine mediate protection against oxidative damage caused by hydrogen peroxide

J. E. Rider; Amy Hacker; Caroline A. Mackintosh; Anthony E. Pegg; Patrick M. Woster; Robert A. Casero

Summary.The polyamines spermidine and spermine have been hypothesized to possess different functions in the protection of DNA from reactive oxygen species. The growth and survival of mouse fibroblasts unable to synthesize spermine were compared to their normal counterparts in their native and polyamine-depleted states in response to oxidative stress. The results of these studies suggest that when present at normal or supraphysiological concentrations, either spermidine or spermine can protect cells from reactive oxygen species. However, when polyamine pools are pharmacologically manipulated to produce cells with low levels of predominately spermine or spermidine, spermine appears to be more effective. Importantly, when cells are depleted of both glutathione and endogenous polyamines, they exhibit increased sensitivity to hydrogen peroxide as compared to glutathione depletion alone, suggesting that polyamines not only play a role in protecting cells from oxidative stress but this role is distinct from that played by glutathione.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis.

Andrew C. Goodwin; Christina E. DeStefano Shields; Shaoguang Wu; David L. Huso; Xinqun Wu; Tracy Murray-Stewart; Amy Hacker-Prietz; Shervin Rabizadeh; Patrick M. Woster; Cynthia L. Sears; Robert A. Casero

It is estimated that the etiology of 20–30% of epithelial cancers is directly associated with inflammation, although the direct molecular events linking inflammation and carcinogenesis are poorly defined. In the context of gastrointestinal disease, the bacterium enterotoxigenic Bacteroides fragilis (ETBF) is a significant source of chronic inflammation and has been implicated as a risk factor for colorectal cancer. Spermine oxidase (SMO) is a polyamine catabolic enzyme that is highly inducible by inflammatory stimuli resulting in increased reactive oxygen species (ROS) and DNA damage. We now demonstrate that purified B. fragilis toxin (BFT) up-regulates SMO in HT29/c1 and T84 colonic epithelial cells, resulting in SMO-dependent generation of ROS and induction of γ-H2A.x, a marker of DNA damage. Further, ETBF-induced colitis in C57BL/6 mice is associated with increased SMO expression and treatment of mice with an inhibitor of polyamine catabolism, N1,N4-bis(2,3-butandienyl)-1,4-butanediamine (MDL 72527), significantly reduces ETBF-induced chronic inflammation and proliferation. Most importantly, in the multiple intestinal neoplasia (Min) mouse model, treatment with MDL 72527 reduces ETBF-induced colon tumorigenesis by 69% (P < 0.001). The results of these studies indicate that SMO is a source of bacteria-induced ROS directly associated with tumorigenesis and could serve as a unique target for chemoprevention.


Journal of Medicinal Chemistry | 2010

(Bis)urea and (Bis)thiourea Inhibitors of Lysine-Specific Demethylase 1 as Epigenetic Modulators

Shiv K. Sharma; Yu Wu; Nora Steinbergs; Michael L. Crowley; Allison S. Hanson; Robert A. Casero; Patrick M. Woster

The recently discovered enzyme lysine-specific demethylase 1 (LSD1) plays an important role in the epigenetic control of gene expression, and aberrant gene silencing secondary to LSD1 overexpression is thought to contribute to the development of cancer. We recently reported a series of (bis)guanidines and (bis)biguanides that are potent inhibitors of LSD1 and induce the re-expression of aberrantly silenced tumor suppressor genes in tumor cells in vitro. We now report a series of isosteric ureas and thioureas that are also potent inhibitors of LSD1. These compounds induce increases in methylation at the histone 3 lysine 4 (H3K4) chromatin mark, a specific target of LSD1, in Calu-6 lung carcinoma cells. In addition, these analogues increase cellular levels of secreted frizzle-related protein (SFRP) 2 and transcription factor GATA4. These compounds represent an important new series of epigenetic modulators with the potential for use as antitumor agents.


Biochemical and Biophysical Research Communications | 2003

Properties of purified recombinant human polyamine oxidase, PAOh1/SMO.

Yanlin Wang; Tracy Murray-Stewart; Wendy Devereux; Amy Hacker; Benjamin Frydman; Patrick M. Woster; Robert A. Casero

The discovery of an inducible oxidase whose apparent substrate preference is spermine indicates that polyamine catabolism is more complex than that originally proposed. To facilitate the study of this enzyme, the purification and characterization of the recombinant human PAOh1/SMO polyamine oxidase are reported. Purified PAOh1/SMO oxidizes both spermine (K(m)=1.6 microM) and N(1)-acetylspermine (K(m)=51 microM), but does not oxidize spermidine. The purified human enzyme also does not oxidize eight representative antitumor polyamine analogues; however, specific oligamine analogues were found to be potent inhibitors of the oxidation of spermine by PAOh1/SMO. The results of these studies are consistent with the hypothesis that PAOh1/SMO represents a new addition to the polyamine metabolic pathway that may represent a new target for antineoplastic drug development.


Expert Reviews in Molecular Medicine | 2013

Polyamines and cancer: implications for chemotherapy and chemoprevention

Shannon L. Nowotarski; Patrick M. Woster; Robert A. Casero

Polyamines are small organic cations that are essential for normal cell growth and development in eukaryotes. Under normal physiological conditions, intracellular polyamine concentrations are tightly regulated through a dynamic network of biosynthetic and catabolic enzymes, and a poorly characterised transport system. This precise regulation ensures that the intracellular concentration of polyamines is maintained within strictly controlled limits. It has frequently been observed that the metabolism of, and the requirement for, polyamines in tumours is frequently dysregulated. Elevated levels of polyamines have been associated with breast, colon, lung, prostate and skin cancers, and altered levels of rate-limiting enzymes in both biosynthesis and catabolism have been observed. Based on these observations and the absolute requirement for polyamines in tumour growth, the polyamine pathway is a rational target for chemoprevention and chemotherapeutics. Here we describe the recent advances made in the polyamine field and focus on the roles of polyamines and polyamine metabolism in neoplasia through a discussion of the current animal models for the polyamine pathway, chemotherapeutic strategies that target the polyamine pathway, chemotherapeutic clinical trials for polyamine pathway-specific drugs and ongoing clinical trials targeting polyamine biosynthesis.


Journal of Medicinal Chemistry | 2009

Recent Advances in the Development of Polyamine Analogues as Antitumor Agents

Robert A. Casero; Patrick M. Woster

Since the publication of our previous Perspective dealing with polyamine drug discovery,1 the field has undergone numerous changes. Most prominent among these changes, from a medicinal chemistry point of view, is that polyamine drug discovery efforts have partially shifted away from an emphasis on inhibitors of polyamine biosynthetic and catabolic enzymes. This shift is in part due to the fact that multiple compensatory mechanisms are available that are able to maintain homeostasis in cellular polyamine pools,2 and thus the utility of specific enzyme inhibitors as drugs is limited. Specific inhibitors are available for every enzyme in the polyamine biosynthetic pathway,1, 3-6 and for the polyamine transport system.7-9 Although these inhibitors have significant effects on their respective target enzymes, only one inhibitor, α-difluoromethylornithine (DFMO) has reached the market. DFMO was originally designed as an antitumor agent, but the drug was not effective enough to warrant continued Phase II trials. However, it has been shown to be an effective cure for infection caused by Trypanosoma brucei gambiense (West African Sleeping Sickness),10, 11 and has recently shown considerable potential as a cancer chemopreventive agent (see below).12-14 Although no other polyamine biosynthesis inhibitor has been advanced to the market, the ubiquitous nature of the natural polyamines would lead one to conclude that these molecules have numerous cellular effector sites that are frequently dysregulated in cancer, and as such should provide a target rich environment for therapeutic intervention. Recent medicinal chemistry efforts in the polyamine field have focused on the discovery of compounds that produce cellular effects that are either independent of, or in addition to the polyamine metabolic enzymes. In addition, polyamine chains have been used to make hybrid drug molecules in order to improve cellular import, increase affinity for chromatin or to serve as carriers. This Perspective will focus on developments in polyamine drug discovery since our previous article.1

Collaboration


Dive into the Patrick M. Woster's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurence J. Marton

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Yi Huang

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Amy Hacker

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tracy Murray-Stewart

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Boobalan Pachaiyappan

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Tracy Murray Stewart

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yanlin Wang

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Anthony E. Pegg

Pennsylvania State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge