Network


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

Hotspot


Dive into the research topics where Thomas E. Wiese is active.

Publication


Featured researches published by Thomas E. Wiese.


PLOS ONE | 2012

Endocrine Disruptor Regulation of MicroRNA Expression in Breast Carcinoma Cells

Syreeta L. Tilghman; Melyssa R. Bratton; H. Chris Segar; Elizabeth C. Martin; Lyndsay V. Rhodes; Meng Li; John A. McLachlan; Thomas E. Wiese; Kenneth P. Nephew; Matthew E. Burow

Background Several environmental agents termed “endocrine disrupting compounds” or EDCs have been reported to bind and activate the estrogen receptor-α (ER). The EDCs DDT and BPA are ubiquitously present in the environment, and DDT and BPA levels in human blood and adipose tissue are detectable in most if not all women and men. ER-mediated biological responses can be regulated at numerous levels, including expression of coding RNAs (mRNAs) and more recently non-coding RNAs (ncRNAs). Of the ncRNAs, microRNAs have emerged as a target of estrogen signaling. Given the important implications of EDC-regulated ER function, we sought to define the effects of BPA and DDT on microRNA regulation and expression levels in estrogen-responsive human breast cancer cells. Methodology/Principal Findings To investigate the cellular effects of DDT and BPA, we used the human MCF-7 breast cancer cell line, which is ER (+) and hormone sensitive. Our results show that DDT and BPA potentiate ER transcriptional activity, resulting in an increased expression of receptor target genes, including progesterone receptor, bcl-2, and trefoil factor 1. Interestingly, a differential increase in expression of Jun and Fas by BPA but not DDT or estrogen was observed. In addition to ER responsive mRNAs, we investigated the ability of DDT and BPA to alter the miRNA profiles in MCF-7 cells. While the EDCs and estrogen similarly altered the expression of multiple microRNAs in MCF-7 cells, including miR-21, differential patterns of microRNA expression were induced by DDT and BPA compared to estrogen. Conclusions/Significance We have shown, for the first time, that BPA and DDT, two well known EDCs, alter the expression profiles of microRNA in MCF-7 breast cancer cells. A better understanding of the molecular mechanisms of these compounds could provide important insight into the role of EDCs in human disease, including breast cancer.


Breast Cancer Research | 2008

Proteomic analysis of tumor necrosis factor-α resistant human breast cancer cells reveals a MEK5/Erk5-mediated epithelial-mesenchymal transition phenotype

Changhua Zhou; Ashley M. Nitschke; Wei Xiong; Qiang Zhang; Yan Tang; Micheal J Bloch; Steven Elliott; Yun Zhu; Lindsey E. Bazzone; David C. Yu; Christopher B. Weldon; Rachel Schiff; John A. McLachlan; Barbara S. Beckman; Thomas E. Wiese; Kenneth P. Nephew; Bin Shan; Matthew E. Burow; Guangdi Wang

IntroductionDespite intensive study of the mechanisms of chemotherapeutic drug resistance in human breast cancer, few reports have systematically investigated the mechanisms that underlie resistance to the chemotherapy-sensitizing agent tumor necrosis factor (TNF)-α. Additionally, the relationship between TNF-α resistance mediated by MEK5/Erk5 signaling and epithelial-mesenchymal transition (EMT), a process associated with promotion of invasion, metastasis, and recurrence in breast cancer, has not previously been investigated.MethodsTo compare differences in the proteome of the TNF-α resistant MCF-7 breast cancer cell line MCF-7-MEK5 (in which TNF-α resistance is mediated by MEK5/Erk5 signaling) and its parental TNF-a sensitive MCF-7 cell line MCF-7-VEC, two-dimensional gel electrophoresis and high performance capillary liquid chromatography coupled with tandem mass spectrometry approaches were used. Differential protein expression was verified at the transcriptional level using RT-PCR assays. An EMT phenotype was confirmed using immunofluorescence staining and gene expression analyses. A short hairpin RNA strategy targeting Erk5 was utilized to investigate the requirement for the MEK/Erk5 pathway in EMT.ResultsProteomic analyses and PCR assays were used to identify and confirm differential expression of proteins. In MCF-7-MEK5 versus MCF-7-VEC cells, vimentin (VIM), glutathione-S-transferase P (GSTP1), and creatine kinase B-type (CKB) were upregulated, and keratin 8 (KRT8), keratin 19 (KRT19) and glutathione-S-transferase Mu 3 (GSTM3) were downregulated. Morphology and immunofluorescence staining for E-cadherin and vimentin revealed an EMT phenotype in the MCF-7-MEK5 cells. Furthermore, EMT regulatory genes SNAI2 (slug), ZEB1 (δ-EF1), and N-cadherin (CDH2) were upregulated, whereas E-cadherin (CDH1) was downregulated in MCF-7-MEK5 cells versus MCF-7-VEC cells. RNA interference targeting of Erk5 reversed MEK5-mediated EMT gene expression.ConclusionsThis study demonstrates that MEK5 over-expression promotes a TNF-α resistance phenotype associated with distinct proteomic changes (upregulation of VIM/vim, GSTP1/gstp1, and CKB/ckb; and downregulation of KRT8/krt8, KRT19/krt19, and GSTM3/gstm3). We further demonstrate that MEK5-mediated progression to an EMT phenotype is dependent upon intact Erk5 and associated with upregulation of SNAI2 and ZEB1 expression.


In Vitro Cellular & Developmental Biology – Plant | 1992

OPTIMIZATION OF ESTROGEN GROWTH RESPONSE IN MCF-7 CELLS

Thomas E. Wiese; Leos G. Kral; Kathleen E. Dennis; W. Barkley Butler; S.C. Brooks

SummaryThe factors involved in estradiol-17β induced growth stimulation of MCF-7 human breast cancer cells have been examined. Wild type MCF-7 cells (and clone E3) were shown to undergo slow growth in phenol-red-free medium containing specific calf sera. The E3 clone was used to document a mean 6-day growth stimulation of 3.35-fold (doubling time=33±3 h) in cultures supplemented with 10−11M estradiol-17β. The serum batch utilized in the culture medium is most important in acquiring significant growth stimulation of MCF-7 cells by estradiol-17β. Regardless of the absence of phenol-red, only selected sera (2 out of 14 tested) supported minimal growth of MCF-7 cells in the absence of added estradiol 17β (doubling time=55±11 h). When a calf-serum-supplemented culture failed to display a complete growth response to estradiol-17β, it was due to the rapid growth of the cells in the control (minus estradiol-17β) flasks. Sera that promoted shorter doubling times for MCF-7 cells cultured in the absence of estradiol-17β were rendered less supportive of growth if treated with dextran-coated charcoal or when cultures were supplemented with the estrogen antagonist ICI 164,384 (10−7M). Pooled extracts of these sera were shown to contain stimulatory levels of estradiol-17β. Dextrancoated charcoal treatment of sera removed or deactivated factors (other than estradiol-17β) which were not only required for the growth of MCF-7 cells, but were necessary for estrogen-stimulated growth. Varying the serum-containing medium, buffer, and nutrient mix or the addition of insulin has no effect on the growth response of these cells to estradiol-17β. These investigations document the culture conditions required to produce a maximal and consistent proliferative effect of E2 on MCF-7 cells without exposing the serum constituent to damaging chemical or absorbent agents.


The Journal of Steroid Biochemistry and Molecular Biology | 1994

Molecular modeling of steroidal estrogens: Novel conformations and their role in biological activity

Thomas E. Wiese; S.C. Brooks

Since the structure and conformation of many estrogenic ligands cannot be described with X-ray crystallographic studies, molecular modeling techniques must be used to generate their 3-dimensional structures. The potential of three molecular modeling methods to simulate the X-ray crystallographic geometry of estradiol-17 beta and various analogs (estratrien-1,17 beta-diol, estratrien-2,17 beta-diol, estratrien-3,11 alpha,17 beta-triol, estratrien-3,11 beta,17 beta-triol, 9 beta-estratrien-3,17 beta-diol-11-one) have been compared. MMP2 molecular mechanics as well as the MOPAC semi-empirical molecular orbital methods, AM1 and PM3, were examined in these studies of estrogens with unique ring distortions. Whereas all three methods were able to simulate reasonable estrogen structures, the MMP2 method was found to reproduce the X-ray geometry of estrogens better than the MOPAC methods. The contribution of crystal packing distortions on the X-ray structures in these comparisons is discussed. Additionally, a molecular modeling dynamics method for the systematic conformational searching of steroidal estrogens is presented. For each estrogen examined, conformational searching produced at least one unique steroid conformation in addition to the X-ray crystallographic geometry. The MMP2 potential energy of predicted conformations and transition barriers of these estrogens has been shown to be less than the free energy of receptor binding. Thus, it is conceivable that estrogen ligands which can exist in a number of conformations may be converted to a preferred geometry by binding within the specific site of receptor. Furthermore, it is suggested that conformational flexibility of estrogens may be an important property of specific ligands for the estrogen receptor.


Journal of Molecular Endocrinology | 2011

Pharmacological inhibition of sphingosine kinase isoforms alters estrogen receptor signaling in human breast cancer

James W. Antoon; William D. Meacham; Melyssa R. Bratton; Evelyn M. Slaughter; Lyndsay V. Rhodes; Hasina Ashe; Thomas E. Wiese; Matthew E. Burow; Barbara S. Beckman

Recently, crosstalk between sphingolipid signaling pathways and steroid hormones has been illuminated as a possible therapeutic target. Sphingosine kinase (SK), the key enzyme metabolizing pro-apoptotic ceramide to pro-survival sphingosine-1-phosphate (S1P), is a promising therapeutic target for solid tumor cancers. In this study, we examined the ability of pharmacological inhibition of S1P formation to block estrogen signaling as a targeted breast cancer therapy. We found that the Sphk1/2 selective inhibitor (SK inhibitor (SKI))-II, blocked breast cancer viability, clonogenic survival and proliferation. Furthermore, SKI-II dose-dependently decreased estrogen-stimulated estrogen response element transcriptional activity and diminished mRNA levels of the estrogen receptor (ER)-regulated genes progesterone receptor and steroid derived factor-1. This inhibitor binds the ER directly in the antagonist ligand-binding domain. Taken together, our results suggest that SKIs have the ability to act as novel ER signaling inhibitors in breast carcinoma.


Journal of Medicinal Chemistry | 2010

Effects of 7-O Substitutions on Estrogenic and Anti-Estrogenic Activities of Daidzein Analogues in MCF-7 Breast Cancer Cells

Quan Jiang; Florastina Payton-Stewart; Steven Elliott; Jennifer L. Driver; Lyndsay V. Rhodes; Qiang Zhang; Shilong Zheng; Deepak Bhatnagar; Stephen M. Boue; Bridgette M. Collins-Burow; Jayalakshmi Sridhar; Cheryl L. Klein Stevens; John A. McLachlan; Thomas E. Wiese; Matthew E. Burow; Guangdi Wang

Daidzein (1) is a natural estrogenic isoflavone. We report here that 1 can be transformed into anti-estrogenic ligands by simple alkyl substitutions of the 7-hydroxyl hydrogen. To test the effect of such structural modifications on the hormonal activities of the resulting compounds, a series of daidzein analogues have been designed and synthesized. When MCF-7 cells were treated with the analogues, those resulting from hydrogen substitution by isopropyl (3d), isobutyl (3f), cyclopentyl (3g), and pyrano- (2) inhibited cell proliferation, estrogen-induced transcriptional activity, and estrogen receptor (ER) regulated progesterone receptor (PgR) gene expression. However, methyl (3a) and ethyl (3b) substitutions of the hydroxyl proton only led to moderate reduction of the estrogenic activities. These results demonstrated the structural requirements for the transformation of daidzein from an ER agonist to an antagonist. The most effective analogue, 2, was found to reduce in vivo estrogen stimulated MCF-7 cell tumorigenesis using a xenograft mouse model.


Experimental Biology and Medicine | 2006

Resibufogenin corrects hypertension in a rat model of human preeclampsia

Hop V. Vu; Monica Ianosi-Irimie; Svitlana Danchuk; Edd Rabon; Toshihiko Nogawa; Yoshiaki Kamano; G. Robert Pettit; Thomas E. Wiese; Jules B. Puschett

The study of the pathogenesis of preeclampsia has been hampered by a relative dearth of animal models. We developed a rat model of preeclampsia in which the excretion of a circulating inhibitor of Na/K ATPase, marinobufagenin (MBG), is elevated. These animals develop hypertension, proteinuria, and intrauterine growth restriction. The administration of a congener of MBG, resibufogenin (RBG), reduces blood pressure to normal in these animals, as is the case when given to pregnant animals rendered hypertensive by the administration of MBG. Studies of Na/K ATPase inhibition by MBG and RBG reveal that these agents are equally effective as inhibitors of the enzyme.


Environmental Health Perspectives | 2012

The organochlorine o,p'-DDT plays a role in coactivator-mediated MAPK crosstalk in MCF-7 breast cancer cells

Melyssa R. Bratton; Daniel E. Frigo; H. Chris Segar; Kenneth P. Nephew; John A. McLachlan; Thomas E. Wiese; Matthew E. Burow

Background: The organochlorine dichlorodiphenyltrichloroethane (DDT), a known estrogen mimic and endocrine disruptor, has been linked to animal and human disorders. However, the detailed mechanism(s) by which DDT affects cellular physiology remains incompletely defined. Objectives: We and others have shown that DDT activates cell-signaling cascades, culminating in the activation of estrogen receptor-dependent and -independent gene expression. Here, we identify a mechanism by which DDT alters cellular signaling and gene expression, independent of the estrogen receptor. Methods: We performed quantitative polymerase chain reaction array analysis of gene expression in MCF-7 breast cancer cells using either estradiol (E2) or o,p´-DDT to identify distinct cellular gene expression responses. To elucidate the mechanisms by which DDT regulates cell signaling, we used molecular and pharmacological techniques. Results: E2 and DDT treatment both altered the expression of many of the genes assayed, but up-regulation of vascular endothelial growth factor A (VEGFA) was observed only after DDT treatment, and this increase was not affected by the pure estrogen receptor α antagonist ICI 182780. Furthermore, DDT increased activation of the HIF-1 response element (HRE), a known enhancer of the VEGFA gene. This DDT-mediated increase in HRE activity was augmented by the coactivator CBP (CREB-binding protein) and was dependent on the p38 pathway. Conclusions: DDT up-regulated the expression of several genes in MCF-7 breast cancer cells that were not altered by treatment with E2, including VEGFA. We propose that this DDT-initiated, ER-independent stimulation of gene expression is due to DDT’s ability to initiate crosstalk between MAPK (mitogen-activated protein kinase) signaling pathways and transcriptional coactivators.


Journal of Medicinal Chemistry | 2011

Biomimetic Syntheses and Antiproliferative Activities of Racemic, Natural (−), and Unnnatural (+) Glyceollin I

Rahul S. Khupse; Jeffrey G. Sarver; Jill A. Trendel; Nicole R. Bearss; Michael D. Reese; Thomas E. Wiese; Stephen M. Boue; Matthew E. Burow; Thomas E. Cleveland; Deepak Bhatnagar; Paul W. Erhardt

A 14-step biomimetic synthetic route to glyceollin I (1.5% overall yield) was developed and deployed to produce the natural enantiomeric form in soy, its unnatural stereoisomer, and a racemic mixture. Enantiomeric excess was assessed by asymmetric NMR shift reagents and chiral HPLC. Antiproliferative effects were measured in human breast, ovarian, and prostate cancer cell lines, with all three chiral forms exhibiting growth inhibition (GI) in the low to mid μM range for all cells. The natural enantiomer, and in some cases the racemate, gave significantly greater GI than the unnatural stereoisomer for estrogen receptor positive (ER(+)) versus ER(-) breast/ovarian cell lines as well as for androgen receptor positive (AR(+)) versus AR(-) prostate cancer cells. Surprisingly, differences between ER(+) and ER(-) cell lines were not altered by media estrogen conditions. These results suggest the antiproliferative mechanism of glyceollin I stereoisomers may be more complicated than strictly ER interactions.


International Journal of Cancer | 2013

20(S)-Protopanaxadiol-aglycone Downregulation of the Full-length and Splice Variants of Androgen Receptor

Bo Cao; Xichun Liu; Jing Li; Shuang Liu; Yanfeng Qi; Zhenggang Xiong; Allen Zhang; Thomas E. Wiese; Xueqi Fu; Jingkai Gu; Paul S. Rennie; Oliver Sartor; Benjamin R. Lee; Clement Ip; Lijuan Zhao; Haitao Zhang; Yan Dong

As a public health problem, prostate cancer engenders huge economic and life‐quality burden. Developing effective chemopreventive regimens to alleviate the burden remains a major challenge. Androgen signaling is vital to the development and progression of prostate cancer. Targeting androgen signaling via blocking the production of the potent ligand dihydrotestosterone has been shown to decrease prostate cancer incidence. However, the potential of increasing the incidence of high‐grade prostate cancers has been a concern. Mechanisms of disease progression after the intervention may include increased expression of androgen receptor (AR) in prostate tissue and expression of the constitutively active AR splice variants (AR‐Vs) lacking the ligand‐binding domain. Thus, novel agents targeting the receptor, preferentially both the full‐length and AR‐Vs, are urgently needed. In the present study, we show that ginsenoside 20(S)‐protopanaxadiol‐aglycone (PPD) effectively downregulates the expression and activity of both the full‐length AR and AR‐Vs. The effects of PPD on AR and AR‐Vs are manifested by an immediate drop in proteins followed by a reduction in transcripts, attributed to PPD induction of proteasome‐mediated degradation and inhibition of the transcription of the AR gene. We further show that although PPD inhibits the growth as well as AR expression and activity in LNCaP xenograft tumors, the morphology and AR expression in normal prostates are not affected. This study is the first to show that PPD suppresses androgen signaling through downregulating both the full‐length AR and AR‐Vs, and provides strong rationale for further developing PPD as a promising agent for the prevention and/or treatment of prostate cancer.

Collaboration


Dive into the Thomas E. Wiese's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guangdi Wang

Xavier University of Louisiana

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas E. Cleveland

Agricultural Research Service

View shared research outputs
Top Co-Authors

Avatar

Shilong Zheng

Xavier University of Louisiana

View shared research outputs
Top Co-Authors

Avatar

Carol H. Carter-Wientjes

United States Department of Agriculture

View shared research outputs
Researchain Logo
Decentralizing Knowledge